996
Views
17
CrossRef citations to date
0
Altmetric
Original Articles

Radiation protection following nuclear power accidents: a survey of putative mechanisms involved in the radioprotective actions of taurine during and after radiation exposure.

Article: 14787 | Received 18 Nov 2011, Accepted 18 Nov 2011, Published online: 01 Feb 2012

Abstract

There are several animal experiments showing that high doses of ionizing radiation lead to strongly enhanced leakage of taurine from damaged cells into the extracellular fluid, followed by enhanced urinary excretion. This radiation-induced taurine depletion can itself have various harmful effects (as will also be the case when taurine depletion is due to other causes, such as alcohol abuse or cancer therapy with cytotoxic drugs), but taurine supplementation has been shown to have radioprotective effects apparently going beyond what might be expected just as a consequence of correcting the harmful consequences of taurine deficiency per se. The mechanisms accounting for the radioprotective effects of taurine are, however, very incompletely understood. In this article an attempt is made to survey various mechanisms that potentially might be involved as parts of the explanation for the overall beneficial effect of high levels of taurine that has been found in experiments with animals or isolated cells exposed to high doses of ionizing radiation. It is proposed that taurine may have radioprotective effects by a combination of several mechanisms: Citation1 during the exposure to ionizing radiation by functioning as an antioxidant, but perhaps more because it counteracts the prooxidant catalytic effect of iron rather than functioning as an important scavenger of harmful molecules itself, Citation2 after the ionizing radiation exposure by helping to reduce the intensity of the post-traumatic inflammatory response, and thus reducing the extent of tissue damage that develops because of severe inflammation rather than as a direct effect of the ionizing radiation per se, Citation3 by functioning as a growth factor helping to enhance the growth rate of leukocytes and leukocyte progenitor cells and perhaps also of other rapidly proliferating cell types, such as enterocyte progenitor cells, which may be important for immunological recovery and perhaps also for rapid repair of various damaged tissues, especially in the intestines, and Citation4 by functioning as an antifibrogenic agent. A detailed discussion is given of possible mechanisms involved both in the antioxidant effects of taurine, in its anti-inflammatory effects and in its role as a growth factor for leukocytes and nerve cells, which might be closely related to its role as an osmolyte important for cellular volume regulation because of the close connection between cell volume regulation and the regulation of protein synthesis as well as cellular protein degradation. While taurine supplementation alone would be expected to exert a therapeutic effect far better than negligible in patients that have been exposed to high doses of ionizing radiation, it may on theoretical grounds be expected that much better results may be obtained by using taurine as part of a multifactorial treatment strategy, where it may interact synergistically with several other nutrients, hormones or other drugs for optimizing antioxidant protection and minimizing harmful posttraumatic inflammatory reactions, while using other nutrients to optimize DNA and tissue repair processes, and using a combination of good diet, immunostimulatory hormones and perhaps other nontoxic immunostimulants (such as beta-glucans) for optimizing the recovery of antiviral and antibacterial immune functions. Similar multifactorial treatment strategies may presumably be helpful in several other disease situations (including severe infectious diseases and severe asthma) as well as for treatment of acute intoxications or acute injuries (both mechanical ones and severe burns) where severely enhanced oxidative and/or nitrative stress and/or too much secretion of vasodilatory neuropeptides from C-fibres are important parts of the pathogenetic mechanisms that may lead to the death of the patient. Some case histories (with discussion of some of those mechanisms that may have been responsible for the observed therapeutic outcome) are given for illustration of the likely validity of these concepts and their relevance both for treatment of severe infections and non-infectious inflammatory diseases such as asthma and rheumatoid arthritis.

Introduction

The strength 9 (Richter scale) earthquake and attending tsunami that recently took place in Japan have led to severe damage to several nuclear reactors. This article has been written with the purpose that it hopefully might be helpful both for those physicians who now have the direct responsibility to take care of patients who have been exposed to high levels of ionizing radiation (while working with the damaged nuclear power plants in an effort to hinder the release of much larger quantities of radionuclides from the reactors), for the health authorities in Japan and for others who participate in international efforts to assist Japan with minimizing the health impact of this severe disaster. Because this survey report has been written in an emergency situation in the hope that the information presented might be useful for Japanese physicians and their patients before it is too late, there has not been enough time available to study all relevant literature in the way an author of a scientific survey article will normally try to do. This means that there very likely will be weak points in this text that might have been discovered if more time had been available for the author – and also more time for criticism of the manuscript by colleagues and critical discussions with them.

However, it is hoped that the author partly might compensate for this weakness by drawing on his experience from several years’ work with partly related (overlapping or parallel) problems in connection with other severe diseases (such as HIV disease and hypervirulent influenza), as well as on his personal experience through several years not only as an allergy patient but also as a patient suffering from chronic poisoning with a substance most likely functioning as a potent reactive oxygen species (ROS)-generating redox cycling agent (like some kind of lipid-soluble analog of the herbicide paraquat), and from successful antidote therapy of this potentially very serious poisoning and associated asthma. Knowing therefore that this survey report is far from perfect, critique from readers will be welcome. However, it is hoped that rather than leading to too much theoretical discussion about moot points (which in the absence of new experimental observations might be more or less futile), this paper may instead function as a stimulus to carry out new experimental studies either in the lab or in form of clinical studies for every important question that cannot yet be settled by observations already available. It is hoped that it may serve not only as a survey of now very well-documented research observations (which, however, are not always utilised practically in clinical medicine as might have been expected, even when from a purely scientific point of view there can not be any reasonable doubt about the validity of the observations), but also as an eye-opener to important questions still imperfectly understood.

The effects of ionizing radiation on taurine excretion and of taurine as a radioprotectant are well documented by animal experiments

There is a good deal of experimental literature (with some of it coming from Japan) about protective effects of taurine and taurine derivatives against biological damage caused by ionizing radiation, but much of this literature is now very old, perhaps so old that it may have been forgotten, and not easily available (if at all) by searching for it on PubMed. Some of the articles are so old that abstracts are not given on PubMed, which means one cannot know what was the outcome of the experiment without studying the original. Sometimes they may also have been published in journals or books not included in this literature data base. As examples of literature demonstrating radiation protection by taurine or taurine derivatives in cultured cells or animals, see references 1-9.

Several studies have shown that taurine excretion is enhanced following exposure to high doses of ionizing radiation Citation10 Citation11 Citation12 Citation13 Citation14 Citation15 Citation16 Citation17 Citation18 Citation19 Citation20 Citation21 , and taurine depletion has also been observed in the blood plasma of human cancer patients after cytotoxic chemotherapy and/or radiation therapy without any change in the blood plasma concentrations of the taurine precursor amino acids methionine and cysteine Citation22. The enhanced urinary excretion of taurine following exposure to high doses of ionizing radiation can most plausibly be explained as a consequence of enhanced passive leakage of taurine out of cells that have been exposed to much ionizing radiation because of damage to their plasma membrane. Something similar may presumably happen during cancer therapy with cytotoxic drugs that often function as potent oxidant stressors, and therefore may cause taurine release not only from those tumor cells which they kill, but from normal host cells as well as a result of damage to their plasma membranes. This article may hopefully help to make it clear why it may be very important for the quality of life and perhaps also survival possibilities of cancer patients that taurine depletion happening as a consequence of rough therapy should be corrected.

While taurine administration before or during irradiation conceivably might have protective effects because taurine functions as an antioxidant protectant, taurine administration afterwards might be beneficial by compensating for taurine losses that have occurred because of the irradiation, thus helping to correct harmful effects of taurine depletion per se. However, it will be shown below that taurine supplementation beyond normal physiological requirements may be beneficial because of the effects both of taurine itself and the taurine metabolites taurine chloramine and taurine bromamine as anti-inflammatory signal substances. Taurine may, moreover, also exert an important antifibrogenic effect.

Evidence showing that taurine functions as a biological antioxidant

Evidence that taurine functions as an important biological antioxidant is in large measure indirect, because taurine has been shown to function as a good antidote against several different toxic substances that have little in common, when considering the whole group of substances concerned, other than acting as oxidant stressors, i.e. as prooxidants, either directly when the substance concerned is a reactive oxidant molecule, indirectly via reactive metabolites (that e.g. might function as ROS-generating redox cycling agents) or because they inhibit important antioxidative enzymes. Such antidote effects of taurine or high capacity for taurine uptake into the cells (or conversely harmful effects of taurine depletion) have i.a. been found against alcohol Citation23 Citation24 Citation25 Citation26 Citation27 Citation28 Citation29 Citation30 Citation31 Citation32 Citation33 Citation34 Citation35 Citation36 Citation37 Citation38 Citation39 Citation40 , against the analgesic and fever-depressing drug acetaminophen (also called paracetamol) Citation41 Citation42 Citation43 Citation44 , against the aminoglycoside antibacterial drug gentamicin Citation45 Citation46 Citation47 , against the immunosuppressant drug cyclosporine A Citation48 Citation49, against the anticancer drugs cisplatin Citation50 Citation51 Citation52 Citation53 Citation54 , doxorubicin (also called adriamycin) Citation55 Citation56 Citation57 Citation58 Citation59 Citation60 Citation61 Citation62 , bleomycin Citation63 Citation64 Citation65 Citation66 Citation67 Citation68 Citation69 Citation70 Citation71 Citation72 Citation73 Citation74 Citation75 Citation76 Citation77 Citation78 , methotrexate Citation79 and taumustine Citation80, against the cytokine interleukin-2 (IL-2), which is used for immunotherapy of cancer Citation81 Citation82 Citation83 Citation84 , against the estrogen receptor modulator tamoxifen, which is used in therapy of breast cancer Citation85 Citation86 Citation87 , against the antiarrhythmic drug amiodarone Citation88, against the beta-adrenergic agonists isoproterenol Citation55 and isoprenaline Citation89 Citation90 Citation91 Citation92 , against nicotine Citation93 Citation94 Citation95 , against oxidized low-density lipoprotein (LDL) Citation96, against oxidized fish oil Citation97, against high glucose levels Citation96 Citation98 Citation99 Citation100 Citation101 Citation102 Citation103 Citation104 Citation105 Citation106 Citation107 Citation108 Citation109 Citation110 , against fructose Citation111 Citation112 Citation113 Citation114 Citation115 Citation116 Citation117 Citation118 Citation119 Citation120 Citation121 Citation122 , against galactose Citation123, against advanced glycation end-products (AGEs) Citation100 Citation124 Citation125, against homocysteine Citation126 Citation127 Citation128 Citation129 Citation130 , against the anti-inflammatory drugs ibuprofen Citation131 and indomethacin Citation132 Citation133, against the herbicide paraquat Citation65 Citation134 Citation135, against carbon tetrachloride Citation136 Citation137 Citation138 Citation139 Citation140 Citation141 Citation142 Citation143 Citation144 Citation145 Citation146 Citation147 Citation148 Citation149 , against menadione Citation150, against 1,4-naphtoquinone Citation137 Citation139, against hydrazine Citation137 Citation139, against trinitrobenzene sulfonic acid Citation151 Citation152, against acrylonitrile Citation153, against perchloroethylene Citation154, against methylene dianiline Citation155, against arsenic Citation156 Citation157 Citation158 Citation159 Citation160 Citation161 Citation162 Citation163 Citation164 , against cadmium Citation165 Citation166 Citation167 Citation168 Citation169 Citation170 Citation171 Citation172 , against mercury Citation173, against lead Citation174 Citation175 Citation176 Citation177 Citation178 Citation179 , against copper Citation180, against iron Citation39 Citation181 Citation182 Citation183 , against ozone Citation184 Citation185 Citation186 Citation187 Citation188 and against NO2 Citation189. The studies on antidote effects of taurine in animals poisoned by bleomycin and amiodarone are of special interest because they show a very important antifibrogenic effect of taurine in the lower respiratory tract, which may be further enhanced by combining taurine with high doses of niacin. This might be very relevant both for treatment of patients with chronic obstructive pulmonary disease (COPD) – in an attempt to delay the progression of this disease – and for prophylaxis of fibrosis in patients who have been exposed to high levels of ionizing radiation.

Taurine has, moreover, also been reported to function as a good scavenger of various aldehydes (when the amino group of the taurine molecule reacts with aldehyde groups) Citation190 Citation191 Citation192 , which might be relevant when large quantities of acetaldehyde are formed following ingestion of too much alcohol, but also in diabetes. It also reacts rapidly with hypohalite ions, such as hypochlorite and hypobromite Citation193 Citation194 Citation195 Citation196 Citation197 , which are formed during inflammatory reactions by haloperoxidases such as myeloperoxidase and eosinophil peroxidase when the respiratory burst of leukocytes such as neutrophils, eosinophils or monocytes is activated. Since there is very abundant literature, which easily can be found by searching on PubMed (simply by using taurine chloramine and taurine bromamine as search words), about the reactions between taurine and hypohalite ions and about the effects of the reaction products, only some few examples are given here. It should be noted that while the reaction products taurine chloramine and taurine bromamine are less reactive than their parent compounds hypochlorite and hypobromite, they are still reactive molecules with oxidant properties; they are absolutely not antioxidants. However, they have anti-inflammatory effects following uptake in human cells Citation198 Citation199 Citation200 Citation201 which happens partly because they inhibit activation of the transcription factor NF-kappaB Citation202 Citation203 Citation204 Citation205 , which is a positive regulator of several different genes with proinflammatory effect Citation206 Citation207 Citation208 .

There are, additionally, also several studies showing that taurine protects (in the same organ or other parts of the body) against damage caused by ischemia and reperfusion in several different organs: in the heart and isolated cardiomyocytes Citation209 Citation210 Citation211 Citation212 Citation213 Citation214 Citation215 Citation216 Citation217 Citation218 Citation219 Citation220 Citation221 Citation222 Citation223 Citation224 Citation225 Citation226 Citation227 Citation228 Citation229 Citation230 , in skeletal muscle or whole limbs Citation231 Citation232 Citation233 Citation234 Citation235 , in the liver Citation236 Citation237 Citation238 Citation239 Citation240 Citation241 Citation242 Citation243 Citation244 Citation245 Citation246 Citation247 , in the kidneys Citation248 Citation249 Citation250 , in the testicles Citation251 Citation252 Citation253 , in the intestines Citation254, in peripheral nerves Citation255, in the lungs Citation256 and in the brain Citation257 Citation258 Citation259 Citation260 Citation261 Citation262 Citation263 Citation264 Citation265 . There are, however, also some reports showing no protective effect of taurine supplementation, or a paradoxical protective effect of poor taurine status, so all literature data are not consistent. However, the reports where a protective effect of taurine was found are far more numerous than those showing no effect or an apparent protective effect of poor taurine status.

It is likely that this protective effect of taurine in ischemia followed by reperfusion may be due to more than one mechanism, and that regulatory effects of intracellular taurine on membrane transport systems for calcium may be part of the reason why taurine is protective in tissues that have been exposed to ischemia followed by reperfusion. But since it is well documented that ischemia followed by reperfusion is attended by strongly enhanced oxidative stress, with only some few examples of the very abundant literature about this being given here for illustration Citation266 Citation267 Citation268 Citation269 Citation270 Citation271 Citation272 Citation273 Citation274 Citation275 , there must be good reason to suspect that the protective effect of taurine against organ damage caused by ischemia and reperfusion may in large measure be explained by its antioxidant effect in living cells. This is supported by abundant experimental data showing that other substances with antioxidative protective effects also have similar protective effects against tissue damage caused by ischemia/reperfusion (not only in the affected organs themselves, but a protective effect was in some studies found also against remote injury in other organs), or that depletion leads to enhanced tissue injury, which has been abundantly demonstrated e.g. for selenium and the selenium-dependent antioxidant protective enzyme glutathione peroxidase Citation272 Citation276 Citation277 Citation278 Citation279 Citation280 Citation281 Citation282 Citation283 Citation284 Citation285 Citation286 Citation287 Citation288 Citation289 Citation290 Citation291 Citation292 Citation293 Citation294 Citation295 Citation296 Citation297 Citation298 Citation299 Citation300 Citation301 Citation302 Citation303 Citation304 Citation305 Citation306 Citation307 Citation308 Citation309 Citation310 Citation311 , for glutathione and glutathione precursors Citation312 Citation313 Citation314 Citation315 Citation316 Citation317 Citation318 Citation319 Citation320 Citation321 Citation322 Citation323 Citation324 Citation325 Citation326 Citation327 Citation328 Citation329 Citation330 Citation331 Citation332 Citation333 Citation334 Citation335 Citation336 Citation337 , for carnosine Citation338 Citation339 Citation340 Citation341 Citation342 Citation343 Citation344 Citation345 Citation346 Citation347 and for melatonin Citation346 Citation348 Citation349 Citation350 Citation351 Citation352 Citation353 Citation354 Citation355 Citation356 Citation357 Citation358 Citation359 Citation360 Citation361 Citation362 Citation363 Citation364 Citation365 Citation366 Citation367 Citation368 Citation369 Citation370 Citation371 Citation372 Citation373 Citation374 Citation375 Citation376 Citation377 Citation378 Citation379 Citation380 Citation381 Citation382 Citation383 Citation384 Citation385 Citation386 Citation387 Citation388 Citation389 Citation390 Citation391 Citation392 Citation393 Citation394 Citation395 Citation396 Citation397 Citation398 Citation399 Citation400 Citation401 Citation402 Citation403 Citation404 Citation405 Citation406 Citation407 Citation408 Citation409 Citation410 Citation411 Citation412 Citation413 Citation414 Citation415 Citation416 Citation417 Citation418 Citation419 Citation420 Citation421 Citation422 Citation423 Citation424 Citation425 Citation426 Citation427 Citation428 Citation429 Citation430 Citation431 Citation432 Citation433 Citation434 Citation435 Citation436 Citation437 Citation438 Citation439 Citation440 Citation441 Citation442 Citation443 Citation444 Citation445 Citation446 Citation447 Citation448 Citation449 Citation450 Citation451 Citation452 Citation453 Citation454 Citation455 Citation456 Citation457 Citation458 Citation459 Citation460 Citation461 Citation462 Citation463 Citation464 Citation465 Citation466 Citation467 Citation468 Citation469 Citation470 Citation471 Citation472 Citation473 Citation474 , but the same also has been reported for various other biological antioxidants. It can also be directly seen from many of the reported experimental studies concerning protective effects of taurine against ischemia/reperfusion damage that this protection must very likely be partly mediated by an antioxidative protective effect, as one can see reduction of the concentrations of direct and/or indirect oxidative stress indicators, compared with the control group, after taurine supplementation.

Since there is much overlap between those chemical mechanisms that cause cellular and organ damage during ischemia followed by reperfusion and those causing cellular damage during exposure to ionizing radiation, with free radicals and other energy-rich oxidant molecules (such as H2O2, peroxynitrite and singlet molecular oxygen) playing a central role not only during ischemia/reperfusion, but also when cells and organs are damaged by ionizing radiation Citation475, there is good reason to believe that most of the experimental data showing protective effects of antioxidant nutrients and the antioxidant hormone melatonin against damage caused by high levels of ionizing radiation also may be relevant for antiischemic protection, and vice versa. The same substances that can protect various organs against damage caused by ischemia and reperfusion may be expected, in a great majority of cases, to be protective against radiation injury as well (which has in several cases also been confirmed by experimental studies), and most substances that can protect against radiation injury would also be expected to have protective effects against tissue damage caused by ischemia and reperfusion.

There is, moreover, much overlap comparing biochemical pathogenetic mechanisms in ischemia/reperfusion and radiation sickness with the biochemical pathogenetic mechanisms in acute or chronic poisoning by substances functioning as strong oxidant stressors (with paraquat and the fungal poison orellanine as typical examples), which explains why several substances that have been found in animal experiments to protect against tissue damage caused by ischemia/reperfusion also have been found to function as good antidotes against several different toxic substances with a prooxidant mechanism of action. There is a very voluminous research literature about this, but most of it is not much practically utilised, not even when the protective substance concerned both is very non-toxic and cheap and presumably can be administrated in high doses with very little risk of serious side effects. Many examples of animal experiments demonstrating such antidote effects can easily be found by combining as search words on PubMed the names of various substances mentioned above for which taurine has been found to function as an antidote (e.g. gentamicin, ethanol or cadmium) with the names of other antioxidant nutrients (e.g. selenium, for which there is an abundant research literature, especially concerning the use of selenium as an antidote against toxic heavy metals and arsenic) or of endogenously produced antioxidants (e.g. melatonin and coenzyme Q10), as well as of polyphenolic plant antioxidants (e.g. silymarin). The present author is himself a chronic poisoning patient who would probably not have been alive without such antidote therapy (having for several years used a combination of high doses of selenium, high doses of coenzyme Q10, high doses of vitamin E and high doses of protein- and taurine-rich fish powder).

Strong parallels may also be found between all these disease situations and what happens during severe inflammatory conditions, both during severe infection (e.g. malignant avian influenza), non-infectious inflammatory diseases including rheumatoid arthritis and asthma, and following severe trauma, e.g. severe burn injuries or mechanical trauma to the brain. During infection, oxidative stress is enhanced partly as a consequence of the use of strong oxidants (such as hypohalite ions and peroxynitrite) as antibacterial and antiviral weapons, and partly because of cytokine-induced enhancement of mitochondrial ROS production. But the same mechanisms leading to enhanced oxidative stress will also be activated during various non-infectious inflammatory conditions, as during allergic inflammation, and also following severe trauma (e.g. severe burn injuries).

For obtaining an optimal therapeutic response, it is probably best in all these cases to use a cocktail of protective substances rather than one substance alone, since there are good theoretical reasons to believe that many of the substances concerned may interact with another in a synergistic fashion, especially when they have completely different biochemical functions or different localization in the cells (e.g. water-soluble versus lipid-soluble antioxidants), or otherwise have completely different mechanisms of action (e.g. free radical scavengers versus redox-active metal ion chelators).

It may also be important to use high enough doses of each substance and take into consideration their pharmacokinetic behaviour, especially for such substances that are rapidly excreted following intake of high doses (such as taurine because of limited capacity for tubular reabsorption in the kidneys) or have a rapid normal turnover because they are rapidly degraded by enzymes (such as melatonin, which must have a rapid turnover because of its role as a hormone participating in the regulation of diurnal rhythms) or by rapid non-enzymatic in vivo oxidation processes (as is probably the case for coenzyme Q10). It might thus be an advantage to use slow-release high-dose preparates both in the case of taurine and melatonin in order to obtain a more prolonged effect following intake of the pill (while for coenzyme Q10, turnover is slow enough that this should not be necessary). Such slow-release high-dose taurine and melatonin preparates are perhaps not commercially available today (certainly not in Norway, but it might perhaps also be difficult to find them in other countries), but they should be easy to make, even for use in acute situations like now following the nuclear accident in Japan.

Should a combination of high-dose melatonin and antioxidant nutrients be used as part of the standard routine for acute therapy of brain stroke, myocardial infarction and similar disorders?

In this article, I have included far more literature references than normal even for a survey article. This is because I believe it may be useful to have too many rather than too few references for those in Japan who in the present emergency situation hopefully might find some of the information I have tried to survey to be useful in their work, when trying to limit as much as possible the health damage caused by the nuclear power plant disaster.

The strength of documentation for a certain biological effect of some given protective substance does not only depend on the quality of individual research reports, but also on their total number as well as on their diversity. When the same effect has been found in several different organs by several different groups of scientists, this is much more compelling evidence than when it has been found only in one organ by one group of scientists. And it is better when not only an overall protective effect of some particular intervention has been demonstrated in a particular organ, but also important parts of the responsible mechanism (or mechanisms) have been explained by experimental observations. One has therefore to be convinced about the validity of the observations, concerning for instance the protective effects against tissue damage caused by ischemia and reperfusion of substances such as taurine, selenium, glutathione and melatonin, by the sheer abundance of good quality research reports, as well as by the number of different organs where similar protective effects have been found, at the same time as much of the mechanisms explaining the protective effects appear to be reasonably well understood.

This is especially important in such cases where something of potential value in practical medicine has been well studied in animal experiments, but still not in human patients in well-conducted clinical trials, with such trials either being few or not existing at all. The laws of physics and chemistry are, nevertheless, the same, regardless of biological species from methanogens to man (and, as far as we can see, everywhere in the observable Universe). And most of the fundamental biochemistry, intracellular signal pathways and extracellular signal substances (including hormones and cytokines) are also the same in all mammalian species, which makes it possible to extrapolate with confidence from much of what has been observed in experiments with one species, e.g. rats, also to other mammalian species, including humans.

One should, of course, be cautious when carrying out such extrapolation and bear in mind those important differences that exist due to different ecological niche adaptations in different species. But with application of experience and good judgement, it is possible to understand the difference between such phenomena that might be different in different species and families of animals because they may be influenced by their different ecological niche adaptations (e.g. when comparing digestive physiology in ruminants and monogastric species, or the immune system in very short-lived mammalian species and long-lived ones) and such phenomena that are so fundamental and universal that they must be expected to be the same everywhere (as e.g. in the case of reactive oxygen species (ROS) reacting with DNA molecules, with ROS scavenger molecules having antimutagenic effect).

It is very strange, when considering how abundant those research data are that show very significant protective effect for some of the substances mentioned here against permanent organ damage caused by ischemia/reperfusion, and how non-toxic and cheap some of them (like taurine, melatonin and glutathione) also are, that the use of these substances has not already become standard routine, e.g. for treatment of stroke patients or patients with myocardial infarction, but also for treatment of other diseases and injuries where permanent organ damage develops as a consequence of ischemia/reperfusion and post-injury inflammatory response (e.g. severe head trauma and drowning). It can hardly be considered knowledge-based or research-based clinical medicine not to utilise data from basal research when they are of so good quality and also so abundant as is demonstrated by the very long literature list of this article, especially for the protective effect of melatonin against damage caused by ischemia and reperfusion in several different organs, but also for taurine.

It is very difficult to find any legitimate scientific reason why this should be so. Rather, the explanation must somehow be at the level of communication (between basal scientists and clinicians), sociology or economy, e.g. in the way clinical research is funded (with too small proportion of the funding of clinical research projects coming from the user and buyer side compared with the proportion of research money coming from the seller side, which means that far too little money is used for clinical research on such substances or methods which could help to make practical medicine not only better, but also much cheaper than it is today). The health economic savings for society if the abundant animal experimental data concerning protection against tissue injury caused by ischemia/reperfusion might be better utilised than now (with much better acute treatment for stroke and myocardial infarction than is common today, e.g. by combining a strong antiischemic protective cocktail with hypothermia) would be potentially enormous, not least for those countries (including Japan), that have a high relative proportion of elderly, when considering the age distribution of the whole population, and where the proportion of elderly can be expected to increase even more during the next decade.

Use of a combination of slow-release, high-dose taurine and melatonin preparates in combination with high-dose coenzyme Q10 and other antioxidants for antidote therapy of acute or chronic poisoning with substances with a prooxidant mechanism of action.

Slow-release high-dose taurine and melatonin preparates might probably be helpful in the future also for treatment of several other diseases and injuries, including acute intoxications with substances having a prooxidant mechanism of action (such as paraquat or the fungal poison orellanine) – in which case it is probably also much better to use a suitable antioxidative antidote cocktail rather than one substance alone.

For treatment of acute poisoning with substances functioning as oxidant stressors, it is presumably best to use a combination both of lipid-soluble and water-soluble antioxidants including melatonin (at a high dosage level) and taurine, as well as coenzyme Q10 for preventing too much disturbance of the energy metabolism of vulnerable target organs (like the kidneys in the case of orellanine poisoning) because of rapid peroxidative coenzyme Q10 degradation. It may also be an advantage for obtaining a rapid therapeutic response in acute situations (e.g. following orellanine ingestion) to rely on high doses of non-toxic substances having a rapid turnover in the human organism, rather than relying too much on such substances that have a much slower turnover and therefore need much more time to build up to new steady state intracellular concentrations.

Possible mechanisms explaining the antioxidative protective effects of taurine

While the reactions between the amino group in the taurine molecule on one side and aldehyde group and hypohalite ions on the other are well documented and easy to understand, these reactions can not alone explain the multiple protective effects of taurine against several different substances with prooxidant action, i.e. against molecules which either themselves are reactive and function as oxidant stressors (such as NO2 and paraquat), which can be metabolized into molecules that function as oxidant stressors, e.g. because an inactive parent substances is converted by hydroxylation to form a ROS-generating redox cycling agent, or can inhibit antioxidative enzymes (which may be an important part of the mechanism of the toxic effect for a number of toxic metals). But reports conflict with another regarding the efficacy of taurine as a scavenger of molecules important as oxidants in living organisms, other than those already mentioned.

Role of taurine as a scavenger of reactive oxidant molecules.

Aruoma et al. reported in 1988 that taurine does not react rapidly with O2 .-, H2O2 or OH., and the product of its reaction with HOCl is still sufficiently oxidizing to inactivate alpha 1-antiproteinase Citation476. They concluded on this background that it seems unlikely that taurine functions as an antioxidant in vivo. By contrast, they found hypotaurine to be a much better antioxidant than taurine Citation476. In a study by another group several years later, taurine's capacity to scavenge peroxynitrite was measured, and it was concluded that taurine is only a weak scavenger of peroxynitrite, and that it does not attenuate sodium nitroprusside toxicity to cells in culture Citation477.

In a more recent study (from 2010), Oliveira et al. found also that taurine does not react with H2O2 Citation478. However, taurine was found to exhibit significant scavenging potential against peroxyl radical, nitric oxide, and superoxide donors Citation478. Their study also evaluated if taurine was able to minimize the in vitro Cu/Zn-superoxide dismutase damage (SOD) induced by peroxynitrite Citation478. Taurine was found to prevent both the formation of nitrotyrosine adducts and the decrease in SOD activity caused by peroxynitrite Citation478. In addition, taurine was found to prevent the ex vivo damage caused by tert-butyl hydroperoxide in rat liver slices Citation478. Oliveira et al. concluded from these experimental data that taurine at different physiological concentrations efficiently scavenges many reactive oxygen and nitrogen species Citation478.

Possible role of taurine as an inhibitor of oxidation reactions caused by perferryl ion complexes and iron-catalyzed reactions between organic molecules and H2O2.

From these partly conflicting reports, one may conclude that taurine is not free of scavenger activity that partly might help to explain its in vivo antioxidant protective effects, but it is not strikingly effective as a scavenger antioxidant except for hypohalite ions and aldehydes. There are many other biological antioxidants that also scavenge reactive molecules, but are more effective than taurine – even though taurine might partly compensate for modest specific effect (at a given concentration) by much higher in vivo concentrations than for many other biological antioxidant molecules.

It should be remembered, however, that it is also possible for a chemical substance to function as a good antioxidant in non-living or living systems without having any scavenger effect at all either for free radicals or for other (non-radical) important oxidant molecules, such as H2O2, peroxynitrite and singlet molecular oxygen. This is because of the crucial role played by iron and other redox-active metals as catalysts of non-enzymatic oxidation reactions both in living organisms Citation479 Citation480 Citation481 Citation482 Citation483 Citation484 Citation485 Citation486 Citation487 Citation488 Citation489 Citation490 Citation491 Citation492 Citation493 and non-living systems (e.g. during storage of foods or animal feeds in contact with air, or in rubber). Substances that can form complexes with such catalytically active metals in a way making them inactive as catalysts will therefore also function as good antioxidants, and can be used for this purpose e.g. in order to enhance the shelf life of various food products.

Citrate is a good example of substances used for this purpose by the food industry. It inactivates the metal atom (hinders it from being catalytically active) by forming a chelate complex, thus hindering reaction between the metal atom and such oxidant molecules that are important during oxidative rancidification, such as molecular oxygen, H2O2, and organic hydroperoxides. At the same time, citrate is also a substance normally formed by human cells, where it is removed as rapidly as it is formed, and completely nontoxic at the levels in which it either is added or can be naturally found in foods.

Taurine, unlike citrate, does not have a structure making it a strong chelator for divalent or trivalent metal ions. But at the high concentrations at which it normally occurs inside living cells, it will instead be capable of forming mixed complexes with iron together with other organic molecules. A type of iron complex that seems to be especially important as a catalytic agent for undesirable spontaneous oxidation processes in living cells are those complexes that can form between iron and organic phosphate molecules, both small ones (such as AMP, ADP, ATP and phospholipids) and macromolecules, especially in form of nucleic acids (DNA and RNA). The reaction between H2O2 and DNA molecules is very sluggish in the absence of redox-active metal atoms that can catalyze the reaction. But this reaction is readily catalyzed both by iron and other redox-active metal ions (including uranium and plutonium, which perhaps might bind even more strongly to DNA compared to iron atoms and hence might be correspondingly more poisonous as catalysts of the reaction between DNA and H2O2), for which reason substances that can bind these metals in a catalytically inactive form are good protectants against H2O2-induced DNA damage Citation494 Citation495 Citation496 Citation497 Citation498 Citation499 Citation500 Citation501 Citation502 Citation503 Citation504 Citation505 Citation506 Citation507 Citation508 Citation509 . There are observations suggesting that redox-active metal ions, especially iron, also are important as catalysts enhancing radiation-induced DNA damage Citation510 Citation511.

Organic phosphate compounds can form complexes, called perferryl ion complexes, where a ferrous iron ion is coordinated to the negatively charged free oxygen atoms in the phosphate group on one side and molecular oxygen on the other Citation512 Citation513 Citation514 Citation515 Citation516 Citation517 . This binding of an oxygen molecule to ferrous iron is similar to what is found in oxyhaemoglobin. But the oxygen molecule is far more reactive in small complexes of this type than it is in the oxyhaemoglobin molecule (perhaps partly for steric reasons??), even though there are some few substances that can react rapidly with oxyhaemoglobin in reactions leading to methaemoglobin formation (with the iron atom in the haemoglobin molecule being oxidized to oxidation number +3).

It should be noted that the coordination environment of the ferrous iron atom before it binds molecular oxygen is completely different for haemoglobin iron and iron attached to phosphate groups in molecules such as ATP. In haemoglobin, the iron atom is coordinated to 4 nitrogen atoms in planar configuration in the porphyrin ring as well as to an imidazole group below, while the iron atom is coordinated to three negatively charged oxygen atoms when sitting on a phosphate group forming a monoester with some organic molecule. The coordination of the iron atom to several nitrogen atoms in oxyhaemoglobin would be expected to lead to greater stability of ferrous compared to ferric iron in the haemoglobin molecule (because of good binding of ferrous iron to nitrogen), while ferric iron would be expected to be relatively more stable when iron is bound to a phosphate group (because of strong electrostatic binding of ferric iron to the negatively charged oxygen atoms). It is plausible to assume that this will cause the oxygen molecule to be more reactive when bound to a ferrous iron atom coordinated to phosphate on the opposite side than when the ferrous iron atom is coordinated to several nitrogen atoms in haemoglobin, since there will be more Gibbs’ free energy reduction when ferrous iron attached to phosphate is oxidized than when the same happens with ferrous iron in the haemoglobin molecule – which would be expected to lead to corresponding energetic facilitation of a simultaneous (and mechanistically coupled?) 1-electron oxidation of an organic molecule when it it is an oxygen molecule attached to ferrous iron with a phosphate group on the opposite side that reacts both with the iron atom and the organic molecule, compared to what happens in reactions leading to methaemoglobin formation.

Small-molecular perferryl ion complexes can form either directly by reaction between the corresponding ferric iron/phosphate complex and superoxide anion radical, or in two reaction steps where the ferric iron first is reduced by some organic reductant, e.g. ascorbate, followed by reaction between the iron atom now in oxidation number +2 and molecular oxygen.

If a ferric iron atom is coordinated to three negatively charged oxygen atoms in a phosphate group, e.g. ATP, and also to three negatively charged oxygen atoms in the sulfonic acid group of taurine, these six oxygen atoms will in combination form an octahedron which will shield the iron atom completely from reaction with any other small molecule, including reactive oxygen species (ROS), organic hydroperoxide groups, organic reductants (such as ascorbate), or molecular oxygen (following reduction of the iron atom to oxidation number +2). In the phosphate group, a highly positively charged phosphorus atom in the centre (oxidation number +5) is surrounded by negatively charged oxygen atoms, with the net charge for the whole phosphate group when it it is coordinated to iron (and not forming a double ester as in DNA and RNA) being -2. In the sulfonic acid group of taurine, a highly positively charged sulphur atom (oxidation number +6) is surrounded by negatively charged oxygen atoms, with the net charge for the whole sulfonic acid group being -1. With two negative charges on the phosphate group, one negative charge on the sulfonic acid group and three positive charges on the iron atom, neutral charge balance is obtained, at the same time as octahedral coordination (6-coordination) is the preferred coordination number both for ferrous and ferric iron when found in silicate and oxide minerals in the Earth's crust Citation518 – for which reason it might be expected that this type of complex might be fairly stable, although far from attaining the stability of chelate complexes, like the ferric iron-citrate complex.

At the same time, it is also reasonable to expect that this type of complex will be sufficiently kinetically labile that it will not hinder the iron atoms from being available for incorporation into iron-dependent enzymes or oxygen-binding proteins such as haemoglobin and myoglobin. Since many of these phosphate-iron-taurine mixed complexes will have low total molecular weight (for the entire complex), much less than for iron-binding proteins such as ferritin and transferrin, they may also be helpful for facilitating the transport of iron atoms by diffusion from one place to another inside the cell (by the principle of facilitated diffusion) in a form where it is not active as a prooxidant catalyst.

In perferryl ion complexes, it is molecular oxygen formally in the same oxidation number as free molecular oxygen which is the reactive oxidizing species. Iron, however, is also very important as a catalyst of reactions involving the reactive oxygen species superoxide anion radical (O2 -.) and hydrogen peroxide (H2O2). These reactions are often referred to as Fenton-type reactions, because it is assumed that hydroxyl radical (OH.) formation by reaction between H2O2 and Fe+ +  is an important part of the reaction mechanism when organic molecules are oxidized. This assumption, although one encounters it almost everywhere in articles where biological effects of H2O2 are discussed, is nevertheless highly doubtful because of the kinetic sluggishness of the Fenton reaction sensu strictu, as far as the rate of hydroxyl group formation (by reaction between H2O2 and Fe+ + ) is concerned (Dr. Erik Løvaas, Tromsø University, personal communication).

A possible alternative might be a more direct reaction, where H2O2 itself reacts both with the organic target molecule and with Fe+ +  that is complex-bound to the same organic molecule, so that the organic target molecule can be oxidized in a 1-electron reaction at the same time as the iron atom is oxidized from ferrous to ferric. A mechanism of this type might perhaps better explain the sequence specificity of DNA lesions Citation495 when DNA reacts in metal-catalyzed reactions with H2O2, compared with the Fenton reaction sensu strictu (i.e. reaction with hydroxyl radical), which because of the very high reactivity of hydroxyl radical might be expected to lead to a more random attack on any potentially reactive part of the DNA molecule (and not only the most reactive groups) that the OH. molecule first will hit-which is poorly compatible with the observed sequence specificity for the DNA lesions.

Mixed complexes between taurine, iron and a phosphate group on the opposite side may, however, be expected to protect also against this type of reaction, whenever phosphate-bound iron is involved in Fenton-type reactions sensu latu Citation519 – although taurine may not necessarily be any good protectant when an iron atom is bound to other groups than phosphate in the DNA molecule.

It is not unlikely that living organisms also may contain other small organic molecules having a similar function as has been proposed for taurine here, either by helping to form mixed complexes with iron in a way making it catalytically inactive, also when it is bound to groups other than phosphate, or by changing the standard redox potential for the ferrous/ferric equilibrium so much that the iron atoms loses most of its prooxidant activity and instead begins to function more as a protective antioxidant e.g. because of superoxide dismutase-like activity of the complex.

Polyamines and trimethylamine oxide (TMAO) might be possible candidates for having an anti-iron protective function in living cells, with the polyamines (because they are already bound to the DNA molecule) perhaps being especially important for protection of the DNA molecule against iron-catalyzed reactions between DNA and H2O2, while TMAO because of its three methyl groups (which conceivably might facilitate binding of the TMAO molecule to the surface of lipid-rich biological membranes) might be suspected of being especially helpful for reducing the rate of iron-catalyzed peroxidation of lipids and protein molecules sitting in biological membranes. The TMAO molecule contains a semipolar bond between the nitrogen and oxygen atoms, conferring a high negative charge density to the oxygen atom that would be expected to facilitate its binding to ferric iron – at the same time as the 3 methyl groups might be expected to provide effective shielding of the iron atom against contact with ROS, hydroperoxide groups or organic reductants when the iron atom is coordinated to the oxygen atom in the TMAO molecule on one side and either to a membrane lipid molecule or a membrane protein on the other side. Perhaps this form of antioxidative protective mechanism for the TMAO molecule might help to explain the high age normally being attained for sharks (that use a combination of urea and TMAO as osmolytes for attaining the same osmolality as seawater) before they die as a direct consequence of old age.

The polyamines are polycations that may be expected competitively to exclude iron atoms from binding to the negatively charged phosphate groups in the backbone of the DNA molecule, rather than forming mixed complexes where the iron atom is sitting between a phosphate group and the polyamine molecule. But it is not inconceivable that polyamines might participate in the formation of mixed complexes with ferrous iron that is simultaneously bound to a nucleotide base on the opposite side in such way that the ferrous iron atom is shielded against contact with molecular oxygen and H2O2.

Without knowing the magnitude of the stability constants of any mixed complex between organic phosphate groups, ferrous or ferric iron and the sulfonic acid group of taurine, it would nevertheless appear plausible to assume that such complexes may be fairly abundant when taurine is found at such high concentrations as are common inside most mammalian cells, with exception of erythrocytes (where the intracellular concentration is about the same as in blood plasma, in great contrast to the situation in the nucleated erythrocytes of fishes that contain much taurine), and that this therefore might be an important mechanism helping to explain why taurine functions as an important intracellular antioxidant Citation520.

It would obviously be desirable to have good measurements of the values for the stability constants of such mixed taurine/organic phosphate complexes both with ferric and ferrous iron. It would also be highly desirable to have more precise information about the stereochemistry of the most abundant complexes between only DNA molecules (and also RNA molecules) and iron, as well as about the stereochemistry of mixed complexes between DNA, iron and taurine (and about the stereochemistry of mixed complexes between RNA, iron and taurine). While there can be little doubt that mixed complexes of this type must be present at much more than negligible concentrations in nucleated mammalian cells (as well as in platelets, which don't have any nucleus, but nevertheless have mitochondria and therefore contain DNA), it is not possible to give any reliable estimate for their quantitative importance as inhibitors of iron-catalyzed oxidation reactions without knowing the stability constants for this type of mixed complexes, combined with what we know about the taurine concentrations in different cell types and different intracellular compartments.

Comparing different types of skeletal muscle in land-living vertebrates, the concentration of taurine is much higher in myoglobin-rich skeletal muscle cells (that also contain much mitochondria), compared to myoglobin-poor muscle cells that contain much fewer mitochondria, but have higher capacity for glycolysis Citation521 Citation522. The myoglobin-poor skeletal muscle cells contain instead much histidine dipeptides Citation522, in humans nearly only carnosine, while in skeletal muscle from poultry both carnosine and anserine are found Citation523. Carnosine is a good antioxidant, but functions also as an important pH buffer and an important anticarbonyl protective agent Citation523. The preferential localization of taurine in such muscle cells where ROS production is higher (i.e. in cells with much myoglobin and several mitochondria), while carnosine is found in such muscle cells where ROS production is normally much lower, might be taken as an argument that taurine may be a more important antioxidant in vivo, compared to carnosine, while carnosine is preferred in such cell types where pH buffering capacity and anticarbonyl protection (because aldehydes, especially methylglyoxal, are formed as byproducts during glycolysis) are relatively more important, compared with antioxidant protection.

It has been proposed that the higher concentration of taurine in myoglobin-rich than in myoglobin-poor skeletal muscle can be explained as a consequence of the localization of much of the taurine inside the mitochondria, where it might possibly also have other functions than antioxidant protection, e.g. pH buffering when pH inside the mitochondrial matrix becomes too high because of rapid extrusion of protons to the intermembrane space by the mitochondrial proton-ATPase Citation522. The taurine concentration may thus be highest in the same parts of the cell where the local H2O2 concentration in contact with DNA molecules is highest, and where it therefore may be especially important to have high concentrations of such other molecules or inorganic ions (e.g. Zn+ +  by competitively excluding iron atoms from binding to especially vulnerable parts of the molecule?) that may help to protect the DNA molecules against damage caused by the iron-catalyzed reaction between DNA and H2O2.

The iron shielding hypothesis presented here is in good qualitative agreement with experimental observations showing that taurine has an antimutagenic effect by protecting DNA molecules against oxidative damage generated by iron-stimulated catecholamine oxidation in the presence of H2O2 Citation524: Calf thymus DNA (100 microg/tube) was exposed to a reaction mixture containing: ferric chloride (60 microM), H2O2 (2.8 mM) and L-dopa (100 microM) Citation524. Taurine and taurine analogs were added simultaneously with H2O2 and L-dopa to determine their efficacies in preventing oxidative damage to DNA. The reaction was carried out for 1 hour at 37 degrees C and terminated by rapid freezing in an ethanol/dry ice bath Citation524. The DNA was precipitated with ethanol and subsequently hydrolyzed with formic acid under vacuum. The hydroxylated bases were separated by HPLC and detected electrochemically. All experiments were replicated a minimum of 5 times Citation524. Taurine (at a concentration of 20 mM) was found to reduce (p < 0.05) damage to DNA as indexed by reductions in the formation of 5-OH-uracil (49% decrease), 8-OH adenine (37% decrease), and 8-OH guanine (21% decrease) Citation524. It had, however, only minimal effects on the formation of 5-OH cytosine (<7% decrease) Citation524. At the same concentration, taurine was also found to increase total DNA recovery after damage by 36-40% and to increase total undamaged guanine by approximately 32% Citation524. It was found that 5-OH-uracil formation also could be reduced (p < 0.05) by the lower concentration of 1 mM taurine and that 8-OH-adenine formation was reduced (p < 0.05) by 5 mM taurine Citation524. The effect of some other substances was also tested, and it was found that total base adduct formation was reduced by 20 mM beta-alanine (30% decreese), by lysine (58% decrease) and even more by glutathione (88% decrease) Citation524. When tested at 20 mM, both hypotaurine and homotaurine provided greater protection against DNA damage than taurine, whereas isethionic acid provided a similar level of protection as taurine Citation524. It may be noted that homotaurine and isethionic acid both contain a sulfonic acid group, like taurine, but homotaurine has greater chain length with 1 carbon atom more than taurine, while isethionic acid has lost the amino group found in taurine and instead contains a hydroxyl group.

The iron shielding hypothesis is also consistent with experimental observations showing that taurine can protect living cells and organisms against toxic effects of iron Citation39 Citation181 Citation182 Citation183 , and that it can inhibit iron catalyzed catecholamine autoxidation in vitro Citation525 Citation526. However, these observations are not sufficient to be taken as proof that the iron shielding hypothesis is correct, and it is not clear if the experimental conditions in the in vitro catecholamine autoxidation experiments were similar enough to conditions in living cells that mixed phosphate-iron-taurine complexes could have been formed as abundantly as normally may happen in various intracellular compartments, when the concentrations of taurine and various organic phosphate compounds simultaneously are high. It is not unreasonable that taurine when the concentration is high enough also can form other iron complexes where the iron is rendered catalytically inactive, e.g. a complex between ferric iron and 2 taurine molecules (where ferric iron is coordinated to 2 sulfonic acid groups instead of one sulfonic acid group plus one phosphate group).

If the iron shielding hypothesis for explaining much of the mechanism of the in vivo antioxidant effect of taurine is largely correct, it means that ascorbate and taurine have complementary roles when they function as antioxidant molecules, since ascorbate is an excellent free radical scavenger Citation527, but it can also have important prooxidant effects Citation528 Citation529 Citation530 , which partly may be explained by its ability to reduce ferric iron to ferrous iron, which is far more reactive than ferric iron both in combination with molecular oxygen Citation512 Citation513 Citation514 Citation515 Citation516 Citation517 and H2O2 Citation519. However, it has been reported that both ferric and ferrous iron must be present if lipid peroxidation shall proceed at a maximal rate Citation531. By shielding the iron atoms in a way preventing their prooxidant catalytic effects, taurine will simultaneously inhibit those prooxidant effects of ascorbate that depend on reactions between ascorbate and iron, but without inhibiting the antioxidant effects of ascorbate because of its function as a free radical scavenger.

Antiinflammatory effects of taurine

Following severe radiation injury, it may be expected that an inflammatory reaction will occur, similarly as happens after other forms of tissue injury (i.a. as a consequence of large release of proinflammatory substances when cells die in necrosis because of the trauma rather than in apoptosis), e.g. because of severe burns (but also following mechanical trauma, e.g. severe head injuries), as well as during infection. While inflammatory reactions take place because they have protective functions normally more important than the harmful ones, too much inflammation can easily be harmful and lead to exacerbation of permanent tissue injury following trauma in one form of another, with a typical example being what normally happens following a brain stroke. It is not at all unreasonable that something similar also can happen in various organs following severe radiation injury (with the post-traumatic inflammation causing enhancement of the risk that the patient shall die e.g. from vascular shock or from multiorgan failure, and also leading to exacerbation of permanent tissue damage if he survives), and that similar methods that have been found in animal experiments to help to reduce an inflammatory over-reaction (and the permanent tissue injury that it causes), e.g. following a brain stroke, also might be valuable for patients suffering from severe injury caused by ionizing radiation.

Is it possible to treat severe inflammation without suppression of antibacterial and antiviral immunity as a side effect?

Since it is common that severe radiation injury also will lead to immunodepression, at the same time as mucosal organs where viruses or harmful bacteria can easily enter and infect the organism also can be injured as a consequence of too much irradiation, the challenge for the patient's doctors will be the same as when one shall try to reduce the risk that a patient will die from hypervirulent avian influenza, viz. to reduce an inflammatory reaction so strong that it is harmful and very dangerous without simultaneously inhibiting too much the organism's immunological defense against viruses and pathogenic bacteria.

What this means is that one must try to weaken so-called neurogenic inflammation (inflammation evoked or strengthened by peripheral nerve cells) and also weaken the activities of such leukocytes that use weapons of so poor specificity that they are capable of inflicting much damage not only to the pathogenic invader, but also to host cells and organs in a way leading to much of what in military jargon is called collateral damage (but in this case to one's own civilian population and not the civilian population of some enemy nation), at the same time as one should also try to strengthen those parts of the immune system that use more precise weapons for targeting the enemy, so that only a minimum of collateral damage to one's own civilian population will ensue. When the enemy soldiers are hiding not in some remote mountain area in Afghanistan, but in the skyscrapers of Manhattan, an American general should obviously stop the over-ambitious colonel who is using machine guns and carpet bombing with dumb cluster bombs killing far more American civilians than enemy soldiers, at the same time as the general might greatly enhance the number of snipers, specially trained commando soldiers and precisely guided drone attacks, as well as trying to do everything else that might be possible for protecting the civilian population. The same principles are also valid if one shall try to save the life of a patient severely affected by hypervirulent H5N5 avian influenza or SARS.

Parallels between the therapeutic challenges in radiation sickness and hypervirulent avian influenza?

I have earlier written some survey articles together with a nutrition scientist from the Norwegian University of Life Sciences and a physicist from the Norwegian Defence Research Establishment, where we discuss how this hopefully may be achieved in cases of hypervirulent influenza (such as H5N1 avian influenza and the Spanish Flu), as well as about the reasons (with obviously important implications for therapy) why the lethality (risk of dying when one is infected) among patients suffering from hypervirulent influenza is strongly influenced both by the nutritional status (with the average surplus mortality being about 10 times higher in British India than it was in Norway in 1918, and also much higher in other poor countries than in those countries that in 1918 were more affluent) and the age of the patient (due to changes in the secretion of various immunostimulatory and immunosuppressive hormones as a function of age, as well as mitochondrial DNA aging) Citation520 Citation532 Citation533.

While much of what is written about therapy in these articles has never been tested on human patients in well-conducted clinical trials, at least not in form of such multifactorial therapeutic interventions as are proposed in our articles, it may be theoretically expected that if such multiple therapeutic interventions can work for H5N1 avian influenza, they can probably work also for a number of other serious infections (e.g. in the hypothetical case that we should get a pandemic with a SARS virus more infective and more lethal than the last one), as well as for patients who have been exposed to high levels of ionizing radiation, when the double challenge is to reduce the level of unspecific harmful post-injury inflammatory response while also trying to reduce the risk that the patient will die from infection as a consequence of the double problem of immunodepression and damage to important mucosal organs.

Taurine can undoubtedly help to improve protection of the “civilian population” (both in the case of hypervirulent influenza and radiation sickness), i.e. reduce the risk that the patient will die from a harmful inflammatory over-reaction, because of its antioxidant effect and capacity for scavenging some of those highly reactive antibacterial and antiviral weapons that have low specificity (are imprecisely targeted) and therefore may be especially dangerous to the “civilians”, such as peroxynitrite and hypohalite ions.

Its effects on immunological functions are, however, ambiguous since taurine on one side has been reported to function as a growth factor for leukocytes and very likely also helps to protect them against some of their own weapons, so that they shall not kill themselves too early instead of killing the enemy – but at the same time, it can also function as a negative regulator of inflammatory and immune reactions by mechanisms that will be explained below, and conceivably might weaken not only harmful inflammatory over-reaction, but also useful antibacterial defense functions (although there are good reasons to believe that it is the former function, i.e. protecting host tissues by hindering harmful immunological over-response, which is the most important one). Taurine has a number of different functions that all may be expected to contribute to an overall anti-inflammatory effect, especially if it can be combined with other substances that have similar effects and a mechanism of action helping them to interact synergistically with taurine (as might possibly be the case – for reasons that will be explained below-with bromide for patients suffering from severe allergic inflammation), giving a multiplicative effect when these other substances are combined with taurine.

Reduction of nociceptive pain and neurogenic inflammation because of reduced prostaglandin production and reduced C-fiber sensitization by oxidatively activated protein kinase C.

First of all should be mentioned that the well-documented in vivo antioxidant effect of taurine (even though the chemical mechanisms explaining it are not completely understood) must be expected to contribute to reduction of prostaglandin synthesis in tissue areas where much damage has occurred, since prostaglandin synthesis is redox-regulated at multiple levels, as has been explained in a recent survey article Citation534. When better taurine status leads to improvement of the antioxidant defense system of our cells, it must be expected that this also will lead to reduction of prostaglandin synthesis in areas of inflammation because of the combination of less protein expression of cyclooxygenase-2 (COX-2), less oxidative activation of phospholipases liberating precursor fatty acids for prostaglandin biosynthesis, and less oxidative activation of the cyclooxygenase Citation534. But reduction of the synthesis of prostaglandins in a tissue affected by inflammation means less prostaglandin sensitization of C-fibres (unmyelinated nerve fibres) in this area Citation534. And less C-fibre sensitization means in turn that the release of proinflammatory neuropeptides such as substance P, neurokinin A and calcitonin gene-related peptide (CGRP) from the C-fibres Citation532 will be reduced. These peptides, because they cause extravasation of plasma proteins through pores that open in the venule walls, are probably important as contributory causes of oedema in the lungs of patients suffering from hypervirulent influenza Citation532. But it is not unreasonable to believe that they might play an important role in diarrhoeal diseases as well.

The sensitivity of C-fibers is also regulated by protein kinase C (PKC), with PKC activation leading to enhancement of the sensitivity of the C-fibers Citation533. They contain several different PKC isozymes that all can be activated by oxidative stress Citation533. There is strong reason to expect that glutathione depletion, e.g. in patients suffering from cancer cachexia, will lead to enhancement of C-fiber sensitivity as a result of enhanced oxidative PKC activation, which might in turn lead to intensification of the patient's pain Citation534. But it is not unreasonable that the activity of PKC in the C-fibers also might be affected by the taurine status of the patient, given the role of taurine as an important intracellular antioxidant.

Since the C-fibers are unmyelinated and very thin (hence having a large specific surface, consistent with their function as chemical multisensors capable of detecting several different abnormalities or normal physiological changes in their local chemical environment), it must be reasonable to suppose that they may have less homeostatic capacity than the brain (which is shielded by the blood-brain barrier) for regulating the intracellular concentrations of nutrient molecules such as taurine, glutathione and ascorbate when the concentrations of these molecules in blood plasma are changed. It is therefore possible that taurine supplementation might help to dampen neurogenic inflammation not only because of reduced prostaglandin synthesis, but also because it may help to reduce oxidative activation of PKC inside the C-fibres.

Another likely consequence of the large specific surface area and lack of myelination in the C-fibres is that they will be much more poorly shielded than cells in the brain to several mutagenic molecules, especially when the latter can act directly as mutagens without prior metabolic transformation (such as ozone in cities with much photochemical air pollutants, acetaldehyde in alcoholics, 4-hydroxynonenal in people eating too much polyunsaturated fatty acids, and inhaled isocyanates, as can happen because of occupational exposure and also happened to a very large number of victims in Bhopal). This may be expected to lead to mutations in the mitochondrial DNA of the C-fibres, which will lead to enhancement of mitochondrial production of reactive oxygen species (ROS) similarly as normal aging does Citation533, with the ROS coming from the mitochondria next causing oxidative PKC activation, which will lead to enhancement of the sensitivity of the C-fibres Citation533 and therefore more pain and more neurogenic inflammation, including enhanced risk of asthma Citation534.

There is a good theoretical rationale (as will become even more apparent from the discussion below about the effects of taurine on inhibitory receptors on the C-fibres) for using taurine as part of a multifactorial therapeutic intervention both for Bhopal victims and other patients suffering from disease caused by too many mutations in C-fibre mitochondrial DNA following exposure to high doses of chemical mutagens from whatever source. Even if many of the Bhopal victims are poor and the number of such patients very large, treatment for reducing the sensitivity of the C-fibres in their lower airways should not need to be overly expensive (too much to make it economically feasible), if one uses a combination of enhanced dietary intakes of selenium and other antioxidant nutrients, enhanced intake of long-chain omega-3 polyunsaturated fatty acids combined with reduction of the intake of omega-6 polyunsaturated fatty acids (especially arachidonic acid if the patient is not too poor to eat much animal foods, but also linoleic acid, being especially important for the poor ones), and high-dose slow-release taurine preparates. Controlled clinical trials for testing the efficacy should, however, first be carried out before applying this form of therapy on the entire affected population in Bhopal.

It would obviously be very useful if similar forms of therapy might be effective also in cities where there is much asthma because of too much air pollution, which is, of course, a major problem in several countries with Mexico City probably being one of the worst examples.

Cellular damage sentinel function of extracellular taurine.

Taurine has multiple physiological roles Citation520 Citation532 Citation535. It is found at much higher normal concentrations (commonly 2 orders of magnitude more) in the cytosol of nucleated cells and platelets than in blood plasma Citation535. Taurine is therefore a suitable candidate for use as a sentinel of plasma membrane damage or disturbance of plasma membrane functions arising e.g. as a consequence of too much oxidative or nitrosative stress, since only a small proportion of the intracellular taurine content leaking out of the cell will be enough to cause a large relative rise in its concentration in the extracellular fluid.

This sentinel function of extracellular taurine can be carried out in two different ways. Extracellular taurine can either bind to receptor sites situated at the outside of the plasma membrane, which may be GABA receptors (which can use either GABA or taurine as ligands) or glycine receptors (which can use either glycine or taurine as ligands), or it can react with reactive halogen species (e.g. hypochlorite or hypobromite) that have been formed by microbicidal, viricidal or parasiticidal peroxidases such as myeloperoxidase, eosinophil peroxidase, lactoperoxidase and female genital tract peroxidase that can catalyze the reaction between halide ions (or thiocyanate) and H2O2 to form anti-inflammatory products such as taurine chloramine and taurine bromamine.

Roles of taurine and glycine as agonist ligands of inhibitory (hyperpolarizing) glycine receptors on macrophages and neutrophils.

Glycine receptors sensitive also to taurine have been found on Kupffer cells, i.e. macrophages in the liver Citation536, where extracellular taurine has an inhibitory action blunting the enhancement of intracellular calcium concentration and TNF-alpha secretion following stimulation of the cells with lipopolysaccharide Citation536. This effect seems to be mediated by a glycine-gated chloride channel Citation536, so that glycine or taurine binding to the receptor will lead to hyperpolarization of the plasma membrane. It has also been reported that production of TNF-alpha and superoxide anion radical (respiratory burst) in alveolar macrophages is blunted by glycine Citation537. Glycine has been shown to have similar inhibitory effects also in other leukocytes, including neutrophils and lymphocytes Citation538. Since taurine is an agonist of the glycine receptor in the macrophages Citation536, it would be expected also to be capable of reducing reactive oxygen species (ROS) production by macrophages both in the lungs, in the liver and in other organs.

Dietary glycine is protective in rat models against tissue damage caused by endotoxemia, liver ischemia-reperfusion, and liver transplantation, and it is believed that this may be mainly explained by glycine inactivating the Kupffer cell via the newly identified glycine-gated chloride channel Citation538. Similarly, it has been reported that taurine (and also betaine) also protects rats from lipopolysaccharide hepatotoxicity as measured by changes in aspartate aminotransferase and alanine aminotransferase activities and total bilirubin levels in serum, and hepatic glutathione contents Citation539. Lipopolysaccharide challenge increased serum TNF-alpha and nitrate/nitrite in rats, which were reduced by betaine or taurine intake Citation539. Glycine-gated chloride channels have, moreover, also been found in the plasma membrane of neutrophils, where glycine similarly has been shown to blunt the respiratory burst Citation540. Taurine should also here be expected to do the same. With neutrophils and monocytes/macrophages both being major players in the pathogenesis of pneumonia induced by hypervirulent influenza, there is good reason to expect that taurine (but also glycine and betaine) might have a similar protective effect in the human lung as has been demonstrated in the liver of experimental animals.

Taurine has also been reported to protect the heart from neutrophil-induced injury during reperfusion following ischemia Citation213. At a concentration of 15 mM, it was found that taurine markedly reduced luminol-dependent chemiluminescence elicited by activated guinea pig neutrophils as well as by chemically generated hypochlorous acid and hydroxyl radicals, but not by chemically generated superoxide radicals Citation213. Even though this scavenging effect of taurine for hypochlorous acid and hydroxyl radicals partly may explain why it was protective during reperfusion following ischemia, it must be reasonable to speculate that this was not the whole mechanism, and that glycine receptor-mediated inhibition of the neutrophils also may have played an important role.

Similarly, it has also been found in various studies Citation244 Citation246 that an important part of the mechanism for the protective effect of taurine against damage caused by reperfusion following warm ischemia in the liver is a blunting of the activity of Kupffer cells (liver macrophages), leading to reduced phagocytosis Citation244, reduced leukocyte/endothelium and platelet/endothelium interactions Citation246 and also being attended by a reduction of the concentration of TNF-alpha in blood plasma Citation244.

Relevance of inhibitory glycine receptors on macrophages and neutrophils for treatment of rheumatoid arthritis.

The glycine receptor-mediated inhibitory effects of taurine and glycine on macrophages and neutrophils might be expected to be therapeutically useful not only for hindering that patients with hypervirulent influenza shall die from asphyxia caused by alveolar oedema Citation532, but also in such non-infectious inflammatory diseases, e.g. rheumatoid arthritis Citation541 Citation542 Citation543 Citation544 Citation545 Citation546 Citation547 Citation548 Citation549 , where macrophages and/or neutrophils are major players in the disease processes leading to tissue destruction. This might perhaps help to explain why the mother of the present author was permanently cured (for nearly 17 years until she died) for an inflammatory disease in the fingers very similar to rheumatoid arthritis after she had started regularly to eat high doses of fish protein concentrate type B (FPC type B), i.e. food-grade fish meal not containing ethoxyquin or other toxic additives, about 50 g per day. FPC type B and fishmeal are good sources both for taurine and glycine, as well as of other substances with anti-inflammatory effects, such as selenium and the long-chain omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) Citation550 Citation551, and it was because of the selenium content my mother started regularly to take high doses. The diagnosis is not entirely certain since mother was never examined by any rheumatology specialist before she was cured, but she had classical textbook symptoms with the basal finger joints being especially severely affected, which led to considerable deformation of the metacarpal bones that persisted after the active inflammatory process had stopped (and could be seen for all the rest of her life until she died).

Possible role of necrotic cell death in the pathogenic mechanism of rheumatoid arthritis.

It is not well understood why so many macrophages and neutrophils accumulate and become activated in the inflamed joints of rheumatoid arthritis patients. However, mitochondrial DNA and oxidized DNA have been detected extracellularly in the synovial fluid of such patients Citation552, which can be taken as evidence suggesting that necrotic cell death may be an important part of the disease process Citation552. It has, however, been reported that mitochondrial DNA can be released from viable neutrophils to form neutrophil extracellular traps, which are extracellular structures able to bind and kill microorganisms Citation553, so one can not totally exclude the possibility that the mitochondrial DNA that has been found extracellularly in the synovial fluid of rheumatoid arthritis patients might partly or entirely have come from this process, rather than necrotic cell death. It may nevertheless be deemed more plausible, at least as a preferred working hypothesis, that necrotic cell death is the most important mechanism leading to liberation of DNA into the synovial fluid of patients suffering from rheumatoid arthritis.

Mitochondrial DNA is a strong proinflammatory agent Citation554 Citation555 Citation556 , acting similarly as bacterial DNA via Toll-like receptors Citation555. Necrotic cell death will occur extensively in affected tissues as a consequence of the disruption of local circulation attending mechanical trauma, and it is also attended by much liberation of other substances with proinflammatory effects Citation556, such as mitochondrially derived formyl peptides Citation556 Citation557 Citation558 , cytochrome c Citation559, and high mobility group box protein-1 (HMGB-1) Citation560 Citation561 Citation562 Citation563 Citation564 (which has a double function both as a nuclear protein and an important cytokine).

Apoptosis is an ATP-dependent process, and necrotic cell death is therefore something that happens when the cell does not contain as much ATP as is needed for apoptosis. An important next question is then what may cause the ATP depletion in cells dying from necrosis in the inflamed joint of a patient suffering from rheumatoid arthritis. It is not implausible that it might be due to a combination of different mechanisms where mitochondrial inhibition by NO Citation565 and cytokines, such as TNF-alpha and IL-1beta Citation566 Citation567 Citation568 , hypoxia, inhibition of mitochondrial enzymes (both in the Krebs’ cycle and the respiratory chain) by too much oxidative stress (e.g. happening partly as a consequence of GSH depletion in the mitochondria) and perhaps also depletion of energy nutrients used by the leukocytes (such as glutamine) all may interact synergistically with each other.

Roles of necrotic cell death and neurogenic inflammation because of excessive C-fibre activation in the pathogenetic mechanisms of lethal systemic responses to extensive tissue injury caused by burns or mechanical trauma: could taurine supplementation be helpful as part of multifactorial treatment strategies for prevention of deaths caused by cardiovascular shock or multiorgan failure following severe trauma?

The extensive liberation of proinflammmatory substances when cells die in necrosis is important for explaining inflammatory responses to injury Citation556. But is not the only causal factor that is important for explaining the post-injury inflammatory response either locally or at a systemic level, since it is also common that there will be much activation of unmyelinated nerve fibres (C-fibres) transmitting pain signals, and when these C-fibres are activated, they release peptides with vasodilatory and proinflammatory effect, such as tachykinins and calcitonin gene-related peptide (CGRP) (which is a topic that we shall return to below).

If the post-injury inflammatory response becomes too strong, it can locally lead to exacerbation of permanent tissue damage (e.g. in form of too much scar tissue development) and at a systemic level to the death of the patient, e.g. from cardiovascular shock or multiorgan failure. When an injured patient feels pain, it happens in large measure as a consequence of much C-fibre activation, and it should not be difficult to understand that too much total liberation of vasodilatory and proinflammatory neuropeptides (when the total mass of injured tissue becomes too large, as in patients with extensive burns) easily may have very harmful consequences at a systemic level either as a consequence of too much reduction of total peripheral arteriolar resistance, making it difficult for the heart to maintain the blood pressure (which might be an important part of the pathogenetic mechanism of cardiovascular shock following extensive trauma), or because of too much inflammation leading to too much oxidative stress not only at the injured sites, but also in other parts of the body (which might be important for explaining the pathogenesis of multiorgan failure).

But the post-injury inflammatory response is not only harmful, since some of the substances released because of necrotic cell death and C-fibre activation are very important as positive signals stimulating antibacterial immunity and subsequent tissue repair. The challenge for the doctor is therefore to hinder that the patient shall die from systemic-level consequences of too strong post-injury inflammatory response (because of too large total liberation of proinflammatory and vasodilatory substances from dead cells and C-fibres), while not suppressing too much either the antibacterial immunity or subsequent tissue repair. This is a double challenge very similar to that also encountered in patients suffering from severe radiation injury or severe infections such as hypervirulent avian influenza or SARS. It is therefore likely that similar treatment strategies (where this double objective can be optimally achieved) might be useful for all these groups of patients – which means it probably would also be useful for clinical researchers working with all these apparently very diverse groups of patients to collaborate much more closely with each other than might be common today and exchange experiences, especially about their successes, but also about their failures when trying interventions that did not lead to the expected positive results.

There is also good reason for hope (as hopefully may be apparent not only from what already has been said, but also from the subsequent discussion), that taurine supplementation might be good for all of the patient groups mentioned above (especially when used as part of a more multifactorial therapeutic intervention) as a method of suppressing inflammation that hopefully will not inhibit antibacterial or antiviral immunity too much, will not hinder subsequent tissue repair and also is reported from animal experiments to be antifibrotic (and thus hopefully might help to prevent excessive scar development at injured sites).

Possible mechanisms of action of fish protein concentrate type B when used as a drug for treatment of rheumatoid arthritis: relevance of glycine receptors on macrophages and neutrophils.

When my mother started to take high doses of fish powder (about 50 g per day), it is possible that there may have been an improvement of mitochondrial ATP production leading to reduction of necrotic cell death in the inflamed tissue because of a combination of several different mechanisms:

My mother was suffering from impaired local circulation in the fingers, causing her fingers to become abnormally cold at low room temperature, but noticed that her fingers became warmer almost immediately after she had taken a large dose of fish powder. The mechanism of this acute effect, which was independently noted by a pharmacologist friend of mine who is now dead (he tried to take a vasodilatory drug for comparison and found that it gave the same effect) and which I can also feel myself, is unknown, but there can be no doubt that there was indeed an improvement of the local circulation in my mother's fingers that preceded recovery from the inflammatory process. It is therefore not at all implausible that improvement of the oxygen supply to her finger joints may have been an important part of the mechanism leading to her recovery.

With less necrotic cell death, there would have been:

  • less liberation of the proinflammatory mediators mitochondrial DNA, formyl peptides, cytochrome c and HMGB1, at the same time as the effect of taurine and glycine from the fish powder on glycine receptors in neutrophils and macrophages would have helped to reduce the proinflammatory effect of these substances after they had been released, with these factors in combination leading to

  • further reduction of the local consumption of O2 by leukocytes and therefore reduction of the severity of local hypoxia, which would have helped to reduce even more the extent of necrotic cell death leading to liberation of mitochondrial DNA, formyl peptides, cytochrome c and HMGB1, as well as to

  • reduction of the production of NO and TNF-alpha in the inflamed tissue, which would have led to further reduction of the degree of inhibition of mitochondrial ATP production by NO and TNF-alpha, which also would have helped to reduce the extent of necrotic cell death leading to liberation of mitochondrial DNA, formyl peptides, cytochrome c and HMGB1.

A positive feedback cycle leading to progressive reduction of the intensity of the disease process might thus have been started, which could explain the process of gradual recovery that happened for my mother over a period of about two or three weeks after the experimental therapy had started, ending with complete cessation of the symptoms of active inflammation and permanent cure.

The reason why this therapy experiment was started was that I had read about antirheumatic effects of selenium in domestic animals in a review article Citation571 and wanted to try this on my mother, with FPC being the most readily available selenium source for me, since no selenium preparates for use in humans were sold in Norway at that time. But I don't think the highly succesful outcome of the experiment can be explained only as caused by the selenium content of the product (at a dosage level corresponding to about 100 micrograms Se per day). I tried afterwards to persuade a rheumatologist to repeat a somewhat modified version of the experiment (using also some other antioxidants in addition to the fish powder) in a more systematic clinical study, explaining to him what I then thought about pathogenetic mechanisms and possible reasons why the therapy I had tried on my mother did work (which was not the same as I believe today could have been the most important mechanisms explaining the therapeutic effect). He was at first enthusiastic about the idea, and continued to be so also later. But it is possible that he may have got cold feet because some of his colleagues were less enthusiastic, and the clinical study we had been talking about was never started (until finally he died).

Taurine and/or glycine supplementation might presumably be therapeutically useful for patients suffering from rheumatoid arthritis because of their at least partly glycine receptor-mediated inhibitory effect both on macrophages Citation244 Citation246 Citation536 and neutrophils and in the case of taurine possibly also because of its antiischemic protective effect Citation209 Citation210 Citation211 Citation212 Citation213 Citation214 Citation215 Citation216 Citation217 Citation218 Citation219 Citation220 Citation221 Citation222 Citation223 Citation224 Citation225 Citation226 Citation227 Citation228 Citation229 Citation230 Citation231 Citation232 Citation233 Citation234 Citation235 Citation236 Citation237 Citation238 Citation239 Citation240 Citation241 Citation242 Citation243 Citation244 Citation245 Citation246 Citation247 Citation248 Citation249 Citation250 Citation251 Citation252 Citation253 Citation254 Citation255 Citation256 Citation257 Citation258 Citation259 Citation260 Citation261 Citation262 Citation263 Citation264 Citation265 helping to reduce the extent of necrotic cell death in the inflamed joints. But because of the pharmacokinetic behaviour of taurine with rapid urinary excretion following intake of high doses, one would presumably need to use a slow-release high-dose taurine preparate for obtaining the desired effect over a sufficiently long period every day.

Possible relevance of glycine receptors on macrophages and the antiischemic protective effect of taurine for explaining the prophylactic effect of taurine against arterial wall degeneration leading to brain stroke in stroke-prone mice and humans.

Another disease where macrophage-induced tissue destruction following severe local hypoxia has been reported to be an important part of the pathogenetic mechanism Citation572, and where it is possible that the antiischemic protective effect Citation209 Citation210 Citation211 Citation212 Citation213 Citation214 Citation215 Citation216 Citation217 Citation218 Citation219 Citation220 Citation221 Citation222 Citation223 Citation224 Citation225 Citation226 Citation227 Citation228 Citation229 Citation230 Citation231 Citation232 Citation233 Citation234 Citation235 Citation236 Citation237 Citation238 Citation239 Citation240 Citation241 Citation242 Citation243 Citation244 Citation245 Citation246 Citation247 Citation248 Citation249 Citation250 Citation251 Citation252 Citation253 Citation254 Citation255 Citation256 Citation257 Citation258 Citation259 Citation260 Citation261 Citation262 Citation263 Citation264 Citation265 of taurine and the glycine receptor-mediated macrophage inhibition by taurine Citation244 Citation246 Citation536 both might be relevant for explaining its protective effect, is cerebral stroke.

It was found in a specially bred strain of spontaneously hypertensive stroke-prone (SHRSP) rats that vascular damage develops due to locally reduced blood supply to the arterial walls in parts of the brain Citation572. Such vascular damage starts (presumably as a result of severe local hypoxia) at the outer layer of the vascular smooth muscle cells located at the furthest site from the vascular lumen Citation572. It leads to activation of macrophages in response to the vascular damage, with this macrophage-mediated inflammatory reaction leading to further progression of the vascular lesions, which in advanced stages of the disease also will affect the inner layers of the arterial wall. This may either lead to vascular wall rupture that causes cerebral hemorrhage or to thrombosis inside the damaged blood vessels that causes cerebral infarction Citation572. The same pathological processes were later confirmed also in human autopsy material Citation572.

Taurine supplementation was found to have a significant protective effect against this disease process in SHRSP rats Citation572. It has been proposed that this can be explained by an antiinflammatory effect of taurine mediated by taurine chloramine functioning as an inhibitor of macrophage activation Citation572. Taurine chloramine is formed by reaction between taurine and hypochlorite, and hypochlorite is formed in a reaction between chloride ions and H2O2 catalyzed by myeloperoxidase. But since myeloperoxidase is expressed by neutrophils Citation573 and monocytes Citation574, while at least in some studies it has been found to be absent from macrophages Citation573, it is far from obvious that the inhibitory effect of taurine on macrophages in the walls of cerebral arteries can be explained as mediated by taurine chloramine.

It is not unreasonable that the glycine-receptor-mediated inhibitory effect of taurine itself on the macrophages Citation536 could be more relevant in this particular context, both in SHRSP rats and humans, at the same time as it is also possible that the antiischemic protective effect Citation209 Citation210 Citation211 Citation212 Citation213 Citation214 Citation215 Citation216 Citation217 Citation218 Citation219 Citation220 Citation221 Citation222 Citation223 Citation224 Citation225 Citation226 Citation227 Citation228 Citation229 Citation230 Citation231 Citation232 Citation233 Citation234 Citation235 Citation236 Citation237 Citation238 Citation239 Citation240 Citation241 Citation242 Citation243 Citation244 Citation245 Citation246 Citation247 Citation248 Citation249 Citation250 Citation251 Citation252 Citation253 Citation254 Citation255 Citation256 Citation257 Citation258 Citation259 Citation260 Citation261 Citation262 Citation263 Citation264 Citation265 of taurine might be relevant for prevention of the hypoxic damage to parts of the arterial wall that precedes macrophage infiltration and activation (and tissue destruction caused by the activated macrophages). If this interpretation is valid, it may be expected that glycine also should have a protective effect (mediated via glycine receptors on the macrophages).

It is, moreover, also possible that other substances than taurine that have been found to have protective effects against tissue damage caused by ischemia and reperfusion in other organs might be protective here as well (because they may help to hinder development of the hypoxic vascular wall lesions that precedes macrophage accumulation and activation). It is therefore possible that glutathione and glutathione precursor amino acids Citation312 Citation313 Citation314 Citation315 Citation316 Citation317 Citation318 Citation319 Citation320 Citation321 Citation322 Citation323 Citation324 Citation325 Citation326 Citation327 Citation328 Citation329 Citation330 Citation331 Citation332 Citation333 Citation334 Citation335 Citation336 Citation337 , selenium Citation276 Citation277 Citation278 Citation279 Citation280 Citation281 Citation282 Citation283 Citation284 Citation285 Citation286 Citation287 Citation288 Citation289 Citation290 Citation291 Citation292 Citation293 Citation294 Citation295 Citation296 Citation297 Citation298 Citation299 Citation300 Citation301 Citation302 Citation303 Citation304 Citation305 Citation306 Citation307 Citation308 Citation309 Citation310 Citation311 , carnosine Citation338 Citation339 Citation340 Citation341 Citation342 Citation343 Citation344 Citation345 Citation346 Citation347 and melatonin Citation346 Citation348 Citation349 Citation350 Citation351 Citation352 Citation353 Citation354 Citation355 Citation356 Citation357 Citation358 Citation359 Citation360 Citation361 Citation362 Citation363 Citation364 Citation365 Citation366 Citation367 Citation368 Citation369 Citation370 Citation371 Citation372 Citation373 Citation374 Citation375 Citation376 Citation377 Citation378 Citation379 Citation380 Citation381 Citation382 Citation383 Citation384 Citation385 Citation386 Citation387 Citation388 Citation389 Citation390 Citation391 Citation392 Citation393 Citation394 Citation395 Citation396 Citation397 Citation398 Citation399 Citation400 Citation401 Citation402 Citation403 Citation404 Citation405 Citation406 Citation407 Citation408 Citation409 Citation410 Citation411 Citation412 Citation413 Citation414 Citation415 Citation416 Citation417 Citation418 Citation419 Citation420 Citation421 Citation422 Citation423 Citation424 Citation425 Citation426 Citation427 Citation428 Citation429 Citation430 Citation431 Citation432 Citation433 Citation434 Citation435 Citation436 Citation437 Citation438 Citation439 Citation440 Citation441 Citation442 Citation443 Citation444 Citation445 Citation446 Citation447 Citation448 Citation449 Citation450 Citation451 Citation452 Citation453 Citation454 Citation455 Citation456 Citation457 Citation458 Citation459 Citation460 Citation461 Citation462 Citation463 Citation464 Citation465 Citation466 Citation467 Citation468 Citation469 Citation470 Citation471 Citation472 Citation473 Citation474 also might be protective.

Roles of taurine and GABA as agonist ligands of inhibitory GABA receptors on C-fibres leading to reduction of nociceptive pain and neurogenic inflammation.

GABAB receptors have been found on peripheral nerve fibres, both on C-fibres and at the end of cholinergic visceral neurons Citation575 Citation576 Citation577 Citation578 . GABAB agonists have inhibitory effects on these nerves, e.g. inhibition of release of substance P from capsaicin-sensitive neurones in the rat trachea Citation578 and of acetylcholine secretion from cholinergic nerves in the lung and in the colon Citation576 Citation577 Citation578 . GABA itself has been shown to inhibit the anaphylactic response in guinea-pig trachea Citation579. It has been found that there are different subtypes of GABAB receptors in the central nervous system which differ in their sensitivity to taurine as an agonist (while all of them, of course, are sensitive to GABA). Since the sensitivity of the peripheral GABAB receptors in relation to taurine (whether it functions as a good agonist or not) has apparently not been studied, and it is not known which subtype of GABAB receptors is expressed there, one can not know for certain whether or not taurine may function as a good inhibitor (acting via the GABAB receptors) in relation to these nerves or not. But it is known from animal experiments that taurine and homotaurine have antinociceptive properties in experimental pain models where pain is elicited either by heating (hot plate, tail immersion and tail flick models) or by low pH (injection of acetic acid into the peritoneum) Citation580 Citation581 Citation582 Citation583 Citation584 .

Anti-asthmatic effects of taurine and taurine-rich fish powder: possible relevance of inhibitory GABAB receptors on C-fibres and parasympathetic cholinergic nerves.

Taurine has also been reported to have beneficial effects in an experimental rat model of asthma, where it significantly reduced the number of eosinophils, the lipid hydroperoxide concentration and the Evans blue dye extravasation in bronchoalveolar lavage fluid Citation585. A therapeutic effect of taurine (when given as an aerosol spray preparation for inhalation) has been reported for human asthma patients as well; it was of comparable magnitude as for commonly used anti-asthmatic drugs Citation586.

All of this is also highly compatible with my own experience, after I for several years have been taking high doses of FPC type B (about 50 g per day), which as already mentioned is rich in taurine, as a drug for self-medication of acute hayfever and asthma. The effect (e.g. reduction of nasal secretion) comes very rapidly, in less than 5-10 minutes, but starts to recede after about 4 hours (which is compatible with what is known about the pharmacokinetics of taurine with rapid urinary excretion when the renal threshold is exceeded).

These observations, when taken in combination, are highly suggestive of an inhibitory effect of taurine on C-fibres, which may in turn lead to reduction of the secretion of proinflammatory peptides such as substance P from unmyelinated peripheral nerves, which means reduction of neurogenic inflammation (even though it could well be possible that there might be more than one pharmacological target explaining the beneficial effect of taurine observed in rats and humans with asthma, so this is not necessarily the only mechanism). It is therefore possible that taurine could be used for reducing C-fibre activity in the lungs and blunting the process of neurogenic inflammation (which possibly might represent an important contributory cause of the alveolar oedema) in patients suffering from pneumonia caused by hypervirulent influenza or SARS. This would be expected not only to lead to reduced extravasation of blood plasma proteins through pores in the venules, as earlier explained, but also less stimulation of leukocytes (i.a. macrophages) by peptides secreted from the C-fibres, as well as reduction of the centrally mediated reflex leading to enhanced ACTH secretion Citation533 and hence enhanced glucocorticoid secretion as a result of enhanced C-fibre activity Citation533.

However, the effect of fish powder (fish protein concentrate type B, or FPC type B for short) in hayfever and asthma is most likely due not to only to its taurine content, since fish may also be a good source of GABA (that serves as an important osmolyte in fish erythrocytes Citation587 and probably also in fish skeletal muscle), and one can not exclude the possibility that the product might also contain other substances with antiallergic effect, e.g. by functioning as a blocker at histamine receptors. It is also possible that the antiasthmatic and anti-hayfever effects of FPC type B may not only be mediated by an inhibitory action on C-fibers, but also by a direct inhibitory effect on leukocytes, since it can not be excluded that not only neutronphils and macrophages, but also eosinophils and/or mast cells may be equipped with inhibitory glycine receptors, which like the corresponding receptors in macrophages might be sensitive both to glycine and taurine. And the protein found in whole fish must be a good source of glycine, not least from collagen in the fish skeleton. FPC type B is, moreover, also a good source of other nutrients Citation550 Citation551 that may be expected to have a more long-term antiinflammatory effect probably relevant for allergic inflammation, such as selenium, glutathione precursor amino acids (plus most likely also glutathione itself, since the product is made from fresh raw material of good quality) and the long-chain omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), with most of the EPA and DHA content being in form of membrane lipids.

Can taurine be used for inhibition of intestinal neurogenic inflammation leading to secretory diarrhoea?

It is not unreasonable that taurine also might have a similar effect in patients suffering from diarrhoea by reducing C-fibre activity in the intestinal mucosa. Substance P is one of the major agents of secretory stimulation in the intestine Citation588 Citation589; it will thus stimulate that secretion of chloride ions, accompanied by sodium ions and water across the mucosal epithelial cells into the intestinal lumen, which is an important part of the pathogenesis of diarrhoeal diseases Citation590. If the secretion of substance P can be reduced by inhibiting the activity of the C-fibres in the intestinal mucosa, this would therefore be expected to help to reduce the chloride secretion and hence the severity of the diarrhoea.

The question might be raised if it is possible for this secretory response to C-fibre activation in the intestine to be anything but harmful to the organism (as is obviously the case when a child dies because of severe dehydration), or stated differently, how it could have helped to enhance the survival probability and thus the Darwinian fitness of our ancestors. One possible answer to this question could be that enhancement of the rate of fluid secretion across epithelial cells lining the surfaces of crypts in the intestinal mucosa may function as a defense strategy for making it more difficult for virus particles to reach the bottom of the crypts and infect the stem cells giving rise to new enterocytes or colonocytes. It is not difficult to understand that it might be useful in a majority of cases if a viral infection can be limited to such cells that even in the absence of infection will only live for a short period before they die and are replaced by new cells, and that it easily might be far worse for the patient if many stem cells also should be infected.

The fluid secretion must then be strong enough to create a current in the central part of the crypt lumen that is faster than the rate of diffusion of virus particles in the upstream direction. This principle can not work just outside the surface of the epithelial cells, where the current velocity in any case must be very low because of fluid adherence to the cell surface (similarly as also happens at the surface of stones lying at the bottom of a swift river). But it might be supplemented by effective killing of viruses in this region, e.g. by peroxynitrite produced just outside the surface of the epithelial cells when NO that has been produced inside the epithelial cells by inducible NO synthase reacts with superoxide anion radical that has been produced by an NAD(P)H oxidase located in their plasma membrane. If the C-fibers could not only stimulate secretion, but also might help to enhance peroxynitrite production at the surface of the epithelial cells, e.g. by helping to enhance epithelial cell expression of inducible NO synthase, they would by doing so orchestrate a good coordinated defense strategy for minimizing the risk that enterocyte or colonocyte stem cells shall be infected by viruses.

Relevance of taurine- and/or GABA-mediated inhibition of C-fibres for treatment of rheumatoid arthritis and other chronic inflammatory diseases

It must, moreover, also be expected that C-fiber inhibition by taurine and/or GABA leading to reduced secretion of proinflammatory peptides from the C-fiber may be therapeutically useful for patients suffering from rheumatoid arthritis, psoriatic arthritis or Bekhterev's disease, since these substances will attract, stimulate or activate (at least in non-human species) several types of leukocytes, including macrophages Citation591 Citation592 Citation593 Citation594 Citation595 Citation596 Citation597 Citation598 Citation599 Citation600 Citation601 Citation602 and neutrophil granulocytes Citation603 Citation604 Citation605 Citation606 Citation607 Citation608 . It is therefore reasonable to believe that the combined C-fiber inhibitory effect of taurine and GABA may have been an important part of the mechanisms explaining the therapeutic effect of high doses of fish powder, when my mother was cured from rheumatoid arthritis.

With reduced release of proinflammatory peptides (tachykinins and CGRP) from the C-fibres, it is reasonable to expect that not only the activity of individual leukocytes, but also their total number in the inflamed tissues would have been reduced. The total rate of oxygen consumption by leukocytes in the inflamed tissues must thus have been reduced (by the combined effect of reduced leukocyte numbers and reduced O2 consumption per cell), which may have helped to reduce the severity of local hypoxia, at the same time as there would also have been a reduction of the production of proinflammatory cytokines, such as TNF-alpha, by leukocytes, again as a consequence both of the reduction of leukocyte numbers and of the activity of individual leukocytes. This would have led to reduction of the inhibitory effect of some of the cytokines themselves on the mitochondrial function of vulnerable target cells (i.e. those cell types that express the appropriate cytokine receptors), at the same time as it would also mean less cytokine stimulation of NO production by other cell types, which means that mitochondrial inhibition by NO also would have been reduced.

At the same time, it must also be expected that a reduction of the release of proinflammatory peptides from the C-fibres must have led to reduction of plasma protein extravasation through pores in the venule walls Citation532, which would mean reduction of oedema and hence reduction of the average diffusion distance for O2 molecules between the capillaries and the sites of O2 consumption by extravascular cells. The rate of O2 diffusion is proportional to the O2 concentration gradient, which is decreased (for a given O2 partial pressure difference along the gradient) when the distance of diffusion is enhanced. A reduction of the distance of diffusion means therefore enhancement of the rate of O2 diffusion from the blood in the capillaries into the surrounding tissue.

This, similarly as for the above-mentioned reduction of the total rate of O2 consumption by leukocytes, would also be expected to lead to reduction of the severity of local hypoxia in the inflamed tissue, with the combined effect of improved tissue oxygenation and reduced extracellular concentrations of NO and proinflammatory cytokines (such as TNF-alpha) being an improvement of the mitochondrial ATP production capacity of leukocytes and synoviocytes which must in turn be expected to lead to reduction of the rate by which cells in the inflamed tissues die by necrosis rather than by apoptosis. This means in turn reduced liberation of the proinflammatory mediators mitochondrial DNA, formyl peptides, cytochrome c and HMGB1 from cells that die by necrosis, and reduced proinflammatory action of these substances – which may presumably interact in synergistic fashion, as far as leukocyte activity is concerned, with the reduced stimulation of their activities by proinflammatory peptides released from the C-fibres because of GABA receptor-mediated C-fibre inhibition.

A reduction of the activity of C-fibres because of reduction of the rate of production of prostaglandins Citation532 Citation533 Citation534 or reduced oxidative activation of protein kinase C in the C-fibres because of improved nutritional status with antioxidant nutrients, such as selenium and glutathione Citation532 Citation533 Citation534 , must be expected to have similar effects as when the activity of the C-fibres is inhibited via GABA receptors, and it is reasonable to expect that all these factors may interact synergistically with each other. The rate of prostaglandin production depends strongly on the fatty acid composition of the diet, being enhanced by a high intake of arachidonic acid and reduced by high intakes of the long-chain omega-3 fatty acids EPA and DHA Citation534. It also depends on antioxidative nutrients such as glutathione and selenium with poor selenium or glutathione status synergizing with high intake of arachidonic acid as causes of enhanced prostaglandin production, which will lead to enhanced neurogenic inflammation Citation534. At the time when my mother was cured, nothing was done to change her intake of arachidonic acid, but since her meat consumption at that time was fairly modest, it is unlikely that her intake of arachidonic acid could have been very high. The fish powder would, however, have helped to enhance her intake not only of antioxidant nutrients, but also of EPA and DHA.

Trying to explain what happened in patients who have been dead for several years can not be regarded as more than working hypotheses that in principle are not possible either to verify or falsify by such methods that can be used for examining patients still alive. The same will also be the case for patients still alive after the disease has been cured. But if the working hypothesis is a biologically plausible one, it should be possible to test it on other patients still alive. Since rheumatoid arthritis is a severe invalidizing disease associated also with excess death rate, and since it is now often treated with drugs having much adverse side effects, including higly mutagenic cytotoxic drugs (which the present author is convinced should not be used at all except for treating life-threatening conditions when no other efficacious therapy is available), the author would like to express his hope that this might be done the sooner the better, even if it is something going far beyond his own capacity and opportunities as a scientist.

Possible roles of high blood sugar level leading to enhanced lactic acid production and vanilloid receptor activation as causes of intensification of nociceptive pain and neurogenic inflammation in patients suffering from rheumatoid arthritis and other chronic inflammatory disorders.

Another factor very important for the regulation of C-fiber activity is the pH value in the intercellular fluid, acting via pH-sensitive activating receptors such as the vanilloid receptor (also called capsaicin receptor because it is activated by capsaicin, which is the active substance in red pepper – Capsicum) Citation534. The rate of lactic acid production in a porly oxygenated tissue must be limited by the availability of carbohydrate that can be metabolized by the glycolytic pathway to form lactic acid Citation534. But the quotient between diffusion rates for glucose and oxygen from the blood in the capillaries into the surrounding tissue is linearly proportional to the glucose concentration in arterial blood plasma Citation534.

It may therefore be expected that a high blood sugar level will lead to enhancement of neurogenic inflammation in patients suffering from rheumatoid arthritis and similar diseases because it leads to enhanced production of lactic acid in the inflamed tissues, which will in turn mean stronger stimulation of the C-fibres via vanilloid receptors. Since I knew nothing about this at the time when my mother was cured (almost 35 years ago), I did not give her any specific advice regarding consumption of carbohydrate-rich foods, which I should certainly have done if I had been aware of it. But she was physically very active in spite of her illness, and she did also eat much vegetables, which are factors that may have helped to reduce her average blood sugar level, in spite of normal consumption of carbohydrate-rich foods such as bread and potatoes.

Antiinflammatory effects of taurine chloramine and taurine bromamine.

Taurine can be halogenated by myeloperoxidase to form taurine chloramine and by myeloperoxidase, eosinophil peroxidase, lactoperoxidase and female genital tract peroxidase to form taurine bromamine, which function as potent anti-inflammatory agents Citation198 Citation199 Citation200 Citation201 Citation609 Citation610 Citation611 Citation612 Citation613 Citation614 Citation615 Citation616 . It is not unreasonable that this also may have been one of the reasons why mother recovered from her rheumatoid arthritis and was completely cured. Taurine chloramine could have been formed because of hypochlorite production by synovial neutrophils and monocytes. I can not reliably estimate the bromide/chloride ratio of her diet, but guess it might have been a little higher than the average for the population in Norway at that time because of abundant intake of vegetable foods, although I don't know if her salt intake was less than average.

Taurine chloramine has been reported to inhibit the production of IL-6 and IL-8 (which is an important chemoattractant for neutrophil granulocytes) by fibroblast-like synoviocytes Citation609 and also to inhibit the proliferation of synoviocytes Citation199 in rheumatoid arthritis. It also inhibits inducible nitric oxide synthase and TNF-alpha gene expression as well as secretion of the chemokines MCP-1 and MIP-2 in alveolar macrophages Citation610 Citation611, which is more directly relevant for patients suffering from hypervirulent avian influenza or SARS, but also may be expected to be highly relevant for patients suffering from rheumatoid arthritis, i.a. because reduction of NO and TNF-alpha production by macrophages in inflamed joint tissues will help to reduce the risk that cells in these tissues shall die from necrosis rather than from apoptosis. The anti-inflammatory effect of taurine chloramine can in part be explained as a consequence of modification of the inhibitory protein IkappaB Citation202 Citation203 Citation204 , which forms a complex with NF-kappaB which must dissociate in order that the transcription factor NF-kappaB shall be activated; taurine chloramine will therefore inhibit NF-kappaB activation Citation202 Citation203 Citation204 Citation610 Citation612 Citation613 Citation616 with taurine bromamine having the same mechanism of action when this substance also inhibits NF-kappaB activation Citation205. For taurine chloramine, it has been shown that this happens because of oxidation of a methionyl group on the IkappaB molecule Citation202.

Taurine chloramine has, moreover, also been reported to inhibit the activation of Ras following lipopolysaccharide treatment of macrophages and to inhibit ERK1/2 activation in a dose-dependent manner in both RAW 264.7 cells and murine peritoneal macrophages, whereas it did not exert any effect on p38 MAPK activation Citation616. It has also other protective effects, since HOCl and TauCl may directly neutralize IL-6 and several metalloproteinases in the extracellular environment Citation615. This may also be expected to be highly relevant in non-infectious inflammatory diseases, such as rheumatoid arthritis, psoriatic arthritis and Bekhterev's disease.

Possible relevance of low bromide concentration in blood plasma leading to reduced production of taurine bromamine by eosinophils in the pathogenesis of asthma.

While myeloperoxidase and possibly female reproductive tract peroxidase are the only mammalian haloperoxidases known to be capable of making hypochlorite from chloride at normal tissue pH values, iodide and bromide have been found to be good reducing substrates Citation617 Citation618 for all mammalian haloperoxidases that have been studied. <l-break/ > These enzymes can have bac-tericidal Citation617, viricidal Citation619 or parasiticidal Citation620 effects, including not only myeloperoxidase, but also lactoperoxidase Citation617, eosinophil peroxidase Citation620 and estrogen-inducible Citation621 female reproductive tract peroxidase. Chloride is also used as substrate for myeloperoxidase, but it is not a good substrate for lactoperoxidase Citation617 and eosinophil peroxidase Citation618, except at low pH. Bromide is normally much more abundant than I- in the blood, but the Br- concentration of blood plasma is most likely strongly dependent on the average Br/Cl ratio of the diet. During precipitation of NaCl from evaporating seawater, there is a strong fractionation of Br- relative to Cl- because of the larger ionic radius of Br- Citation622 compared to Cl-. This may explain why the Br/Cl ratio of table salt analysed in Finland Citation623 was only 1/27 of the Br/Cl ratio found in seawater Citation622.

In marine aerosols, the Br/Cl ratio is higher than in seawater because of evaporation of biologically formed CH3Br from the sea surface Citation624. The average Br/Cl ratio of rain falling over the continents is therefore higher than in seawater, leading to fairly high Br concentrations in terrestrial plants Citation623. But there is reason to expect that a large intake of Br-poor table salt may lead to strong reduction of the average Br/Cl ratio of the total diet and most likely also a corresponding reduction of the Br- concentration of blood plasma and other body fluids. It must be expected that depletion of soil Br (and I) as a consequence of deforestation will lead to further exacerbation of this problem.

There are reasons to believe that blood plasma Br- depletion will lead to impaired killing efficiency for all of the microbicidal peroxidases, but more so for lactoperoxidase and eosinophil peroxidase than for myeloperoxidase. Possible consequences might be enhanced risk of mastitis in lactating women (which will enhance the risk of vertical transmission of HIV from mother to child) as well as in dairy cows and goats, impaired killing of tubercle bacilli, and impaired killing of HIV in the vagina before the virus has managed to infect dendritic cells or other leukocytes. However, the kinetic properties of female reproductive tract peroxidase with different halide substrates have apparently never been adequately studied.

In the most common form of allergic asthma Citation625 Citation626 Citation627 Citation628 , as well as in other allergic inflammation, eosinophils are major players. In asthma and other allergic diseases, it is therefore the products formed by reaction between taurine and such hypohalite ions that are formed by eosinophil peroxidase that might be most important as anti-inflammatory mediators. Since chloride is of very little importance here, and the production of hypobromite by the eosinophil peroxidase reaction must depend strongly on the bromide concentration in the blood, it may be expected that the reduction of plasma bromide concentrations happening as a consequence of the ingestion of much ordinary table salt may be an important cause of enhanced disease activity (because of reduced production of the inhibitor taurine bromamine) in asthma and other allergic diseases. It may be theoretically expected that a combination of taurine supplementation with normalization of the bromide concentration in blood plasma to the same level as is commonly found in mammals when living in their natural environments might have substantial therapeutic effect by helping to maximize the production of anti-inflammatory taurine bromamine in tissue areas affected by allergic inflammation with eosinophil accumulation and activation.

Proinflammatory effects of taurine and magnesium depletion and enhanced oxidative stress because of taurine-mediated, magnesium-mediated and oxidative stress-mediated modulation of calcium concentrations in the cytosol of cells participating in inflammatory reactions?

Finally, it is also possible that taurine might exert an anti-inflammatory effect because of the effects of intracellular taurine on calcium transport across various cellular membrane structures Citation535, most likely because taurine functions as a positive allosteric regulator of membrane electrolyte pumps with only weak binding to the allosteric regulatory binding sites. Since magnesium is needed for the activity of membrane ATPases and intracellular magnesium depletion leads to inhibition of Na+/K+-ATPase and Ca+ + -ATPase activities, taurine depletion will have similar consequences as magnesium depletion for calcium concentrations in the cytosol. The Na+/K+-ATPase Citation629 Citation630 Citation631 and Ca+ + -ATPase Citation632 Citation633 Citation634 are, moreover, both inhibited by oxidative stress and peroxynitrite, which means impaired transport of calcium of away from the cytosol. Cellular depletion of substances important for normal antinitrative or antioxidative defense, such as glutathione or taurine, may therefore be expected to lead to exacerbation of this problem.

But calcium is very important for controlling the discharge of secretory granula into the extracellular fluid. This is also the case when the contents of secretory granula containing substances with proinflammatory effect are released during a disease process attended by inflammation, as exemplified by the release of histamine from mast cells during an allergic reaction. During allergic inflammation, it may, moreover, also be significant that 5-lipoxygenase, which is a rate-controlling enzyme in the leukotriene biosynthesis pathway, is positively regulated by calcium Citation635. If radiation injury is severe enough to be attended by significant simultaneous losses of intracellular taurine, potassium and magnesium, it may be expected that all these factors may tend to enhance the calcium concentration in the cytosol of leukocytes that can discharge secretory granules containing histamine or other proinflammatory substances at the same time as leukotriene synthesis also might be enhanced.

For correction of these problems, it must obviously be important to try to normalize simultaneously both the intracellular K+ and Mg+ +  concentrations, intracellular taurine and intracellular concentrations of glutathione and other water-soluble and lipid-soluble antioxidants, such as ascorbate and ubiquinol (coenzyme Q10 in its reduced form).

Role of ascorbate as reducing cofactor for enzymes scavenging H2O2 and of coenzyme Q10 as an antioxidant.

Ascorbate is important for antioxidant defense not only because of its function as a water-soluble free radical scavenger Citation527, but also because it functions as reducing substrate for the H2O2- and organic hydroperoxide-scavenging Citation636 Citation637 enzyme 1-Cys peroxiredoxin (also called peroxiredoxin-6) Citation636 Citation637 and for the H2O2- scavenging enzyme ascorbate peroxidase. Peroxiredoxin-6 does not only use ascorbate Citation638, but can also use reduced glutathione (GSH) Citation636 Citation637 and dihydrolipoate Citation639 as reducing cofactors. Peroxiredoxin-6 is expressed Citation640 very amply Citation641 in the lungs, and it is also expressed in the mucosa of the mammalian colon Citation642 Citation643, in the kidneys Citation640 Citation644, in the testicles Citation645, in oocytes Citation646 and ovarian cumulus cells Citation646, and in other parts of the brain Citation647 Citation648. Many of the reports about this enzyme are, however, from non-human mammalian species, and it is possible that its organ distribution has still not been well enough studied in humans.

Ascorbate peroxidases are heme proteins that have been much better studied in cyanobacteria Citation649, algae Citation650 and plants Citation651 than in animals. However, an ascorbate peroxidase has been found also in the mammalian eye and choroid plexus Citation652 Citation653. Its distribution in the rest of the human body, e.g. whether it is expressed in the brain or not, seems to be unknown. Coenzyme Q10, especially in its reduced form ubiquinol, is an important endogenously synthesized lipid-soluble antioxidant Citation654 Citation655 Citation656 Citation657 Citation658 , which because of its location in biological membranes conceivably might be especially important for protection not only of the membrane lipids, but also of membrane proteins against oxidative damage.

The anti-inflammatory and analgesic effects of taurine could be relevant in a wide range of disease situations ranging from highly lethal pulmonary infections to common pain.

The anti-inflammatory and analgesic effects of taurine might presumably be clinically relevant in a very wide range of disease situations (including hypervirulent avian influenza, SARS, diarrhoeal diseases, rheumatoid arthritis, Bekhterev's disease, asthma, painful cancer. common muscle pains such as tension headache, and may be also menstrual pain and pain associated with childbirth), as well as following severe injuries, e.g. severe burn injury (in which case there is good reason to suspect that too much neurogenic inflammation with too large total release of tachykinins and CGRP may be a very important part of the pathogenetic mechanism leading to the death of severely injured patients).

Used as an analgesic, taurine has the important advantage that it is almost completely non-toxic and free of potentially dangerous side effects, except for patients with type 1 diabetes (because of a hypoglycaemic effect that possibly might be explained by taurine-induced enhancement of the sensitivity to insulin following its binding to the insulin receptor Citation659 Citation660 Citation661 , and which might conceivably lead to the development of potentially dangerous hypoglycaemia following insulin injection). This is in great contrast to all currently used NSAIDs, which all can have important side effects (e.g. in form of severe gastric hemorrhage induced by aspirin), and also paracetamol, which is mutagenic Citation534 and greatly enhances the risk of asthma Citation534, which is a long-term and most likely irreversible side effect that possibly might be related to the mutagenic effect of this drug because of damage to the mitochondrial DNA in C-fibres and other cells in the lower airways, which could lead to enhanced C-fiber sensitivity (because of enhanced oxidative activation of protein kinase C) and in other cell types could lead to enhanced expression of various proinflammatory genes (because of enhanced activity of oxidatively regulated transcription factors, such as NF-kappaB) Citation534.

But if taurine shall be commonly used as an analgesic as as an anti-inflammatory drug, e.g. for treatment of asthma (in combination with normalization of the blood plasma concentration of bromide to the same level as is commonly found in wild animals), its rapid renal excretion is a problem, and it would probably be much better if slow-release taurine formulations at a suitable dosage level had been commercially available, rather than using taurine pills or capsules with rapid intestinal absorption.

Clinical experience using taurine or taurine-rich fish powder for treatment of severe infections (tuberculosis or measles) attended by diarrhoea

There is a very interesting report from the 1930s from a group of Japanese medical scientists (H. Sugihara, S. Nagasawa and H. Okabe) who had studied the effects of taurine both in experimental animals and human patients Citation662. The report was published in German language in a medical journal in Germany (Klinische Wochenschrift), and it is not unreasonable that it may have been overlooked, despite the importance of the reported observations, and later may have become completely forgotten in English-speaking countries, including the United Kingdom and the United States, both as a result of the political situation during the 1930s and World War 2 (which was still remembered in UK and the United States after World War 2 was over, including those serious war crimes that had been committed by medical professionals as part of the genocidal acts of the Hitler regime, especially when carrying out cruel medical experiments on human subjects) and because of the language barrier between German-speaking and English-speaking countries. But this article has nothing to do with those war crimes for which medical professionals both from Germany and Japan were responsible during the 1930s and 1940s; there is absolutely nothing ethically objectionable in it, and it is still well worth reading.

The observations, especially regarding taurine effects on the heart, that Sugihara, Nagasawa and Okabe report from their studies on experimental animals are in good agreement with what other scientists have found in studies on other animal species that have been carried out more than 30 years later. For this reason, it is reasonable to believe that their reports about clinical observations on human patients also may be similarly accurate and trustworthy, even in such cases where the observations have not been independently checked for verification or falsification by other groups of scientists.

After Sugihara et al. had first observed taurine effects on nerve reflexes in animal experiments, suggesting it might have an analgesic effect, they tried to find out if it might also have a similar effect on human patients. They gave taurine in form of intravenous injections at a very high daily dosage level (from 500 mg to several grams per day) to adult human patients suffering from pulmonary tuberculosis and also chronic diarrhoea with severe abdominal pain, most likely as a consequence of intestinal tuberculosis. Following the taurine injection, several of the patients responded by very rapid pain relief, as illustrated by excerpts from the patient journals quoted in the article Citation662. This also happened in patients who were resistant to the analgesic effect of opiates, but obtained very rapid pain relief following the parenteral administration of taurine. When taurine was given every day, it also had an antidiarrhoeal effect in several patients. But this effect did not come immediately, as with the pain relief, but developed more gradually over some days, perhaps up to about one week after the treatment with taurine had started.

Sugihara et al. report, moreover, also about peripheral administration of taurine, especially for treatment of painful ulcerations (decubitus), where a 0.5% aqueous taurine solution was used, which was applied locally over the surface of the painful ulceration. This did not lead to immediate pain relief as when taurine was given parenterally, but over may be some few hours, excellent pain relief was observed also here. The rapid effect of taurine following intravenous administration and its effect also when applied peripherally must both be regarded as strong evidence suggesting a peripheral rather than a central nervous mechanism for the analgesic effect. These observations by Sugihara et al. are also in good agreement with those animal experiments performed several years later that have demonstrated analgesic effects of taurine and/or homotaurine against pain induced either by hyperthermia (tail flick and tail immersion experimental models) or injection of acetic acid in the peritoneum Citation580 Citation581 Citation582 Citation583 Citation584 .

It is not unreasonable that the antidiarrhoeal effect of high doses of taurine following intravenous injection that was also reported by Sugihara et al. partly might be explained by an anti-inflammatory effect of taurine in the intestine, which might be mediated by a combination of various mechanisms discussed above. However, the time curve for the antidiarrhoeal effect is not easily explained by an anti-inflammatory effect alone. It would be easier to explain the delayed therapeutic response if it might be postulated that it also may have depended on repair of a damaged intestinal mucosa, where reduction of severe inflammation (which might have happened almost immediately after taurine therapy was started) possibly may have been required before the repair process could start, but where it is also possible that taurine may have functioned as a growth factor facilitating rapid growth of enterocyte precursor cells (for reasons that will be explained below) and hence the repair process.

The observations of Sugihara et al. are in good agreement with my own observations from treatment of a large number of patients suffering from the combination of malnutrition, measles and diarrhoea when I was working for the Red Cross in Niger in 1974 Citation663. About 150 patients were suffering from the combination of malnutrition and measles, and roughly 1/3 of them were suffering from diarrhoea as well. Patients without diarrhoea were given porridge made from a combination of African millet (Pennisetum sp.) and fish protein concentrate, type B (FPC type B), i.e. food-grade fish meal, that had been produced from capelin (Mallotus villosus) that had been caught during the winter season before spawning outside the coast of Finnmark, North Norway. Like most other products made from whole marine fish, not to speak of marine invertebrates such as mussels or squid, FPC made from capelin is an excellent source of taurine.

The porridge was made from 1 part (by weight) FPC and 10 parts unrefined millet flour. The patients were often given extra FPC in pure form, and received also reconstituted milk from skimmed milk powder that had been fermented because of the problem of lactose intolerance, as well as vitamin A in form of red unrefined palm oil (which is an excellent source of beta-carotene, i.e. provitamin A), and vitamin A tablets for those who had symptoms of vitamin A deficiency. The diarrhoea patients received less porridge than those without diarrhoea, but more fishpowder so that the protein intake should be similar in both groups, and also oral rehydration solution (which was just homemade half-isotonic NaCl solution) and ample well-fermented milk (so as to get rid of as much of the lactose content of the milk powder as possible).

It should be noted that fish protein concentrate apart from its protein (which is of excellent quality in all fish products of good quality) at a concentration of about 70% (of total weight) has a much better composition than myoglobin-poor fish fillet products, such as salmon fillet and cod fillet Citation550 Citation551 Citation623, because of the much higher concentrations of several important nutrients in the fish viscera and skeleton compared to the fish muscle Citation551, and its mineral nutrient composition is also better than for dairy products and light-coloured meats, such as poultry meat and pork meat Citation550 Citation551 Citation623. It also complements a cereal-based diet as a source of several nutrients that are found at much lower concentrations, are moore poorly available for intestinal absorption or absent in cereals, but which are more typically associated with or better available from animal foods (such as vitamin B12, taurine, iron, zinc, calcium, some of the essential amino acids and probably carnitine) or more typically associated with seafoods (such as iodine, selenium, vanadium, long-chain omega-3 fatty acids and again taurine).

It should also be noted that cow's milk is a very good source of the intracellular electrolytes potassium, magnesium and phosphorus Citation623, calculated per g protein in the milk (with the K/protein and Mg/protein ratios substantially higher than in FPC Citation550 Citation551 Citation623 and most other animal foods), which might be important for many patients suffering from the combination of protein-energy malnutrition and diarrhoea, but cow's milk contains very little taurine. Milk is also a better source of riboflavine than fish powder. The concentrations of nucleotides/nucleosides, calculated relative to protein concentrations, were most likely much higher in the fish powder than in unfermented cow's milk, which is not a good source of nucleotides/nucleosides, compared to human milk Citation664 Citation665. The fish powder had been produced from capelin (Mallotus villosus) that had been caught in the winter during the spawning season. It must therefore have contained much DNA, especially from the gonads of the male fish. It is likely, however, that the nucleotide/protein ratio of cow's milk may be much enhanced during fermentation because of bacterial synthesis of RNA and DNA. The total nucleotide intake (both from the fish powder and the fermented milk) must therefore have been high for my African patients, which is likely to have been helpful both for the recovery of their immune system Citation664 and for rapid recovery of their intestinal epithelium Citation664.

Before treatment was started, the death rate had been high; we were told that at the average 2 to 3 patients had been dying every day, and some days up to 5 or 6 patients. But after the combined therapy with oral rehydration and refeeding had started (plus antiinfectious drugs for those who had symptoms of other infectious diseases needing drug therapy), the deaths stopped immediately, and most of the diarrhoeas stopped in only some few days. It is difficult for me to explain the dramatically rapid response to therapy in these patients, unless there has been a very rapid repair of the intestinal mucosa of the diarrhoea patients at the same time as there must have been a very rapid recovery of immunological functions. This should not be impossible, when taking into consideration the very high rate of cellular growth both in the precursor cells giving rise to mature enterocytes and among various classes of leukocytes and leukocyte progenitors, when malnutrition is corrected so that these cells can be supplied with an abundant supply of all nutrients needed for their growth, both essential and conditionally essential ones (such as nucleotides and taurine).

I suspect that the clinical observations of Sugihara et al. from treatment of tuberculosis patients with chronic diarrhoea and my own observations from treatment of patients suffering from the combination of malnutrition, measles and diarrhoea in Africa both might be relevant for treatment of patients suffering from substantial gastrointestinal symptoms because of radiation injury. The doctor should not be tempted to resignate too early when faced with an apparently hopeless situation, following severe radiation injury. And it might presumably be easier both for the doctor and the patient to have some hope, when focussing more on what radiation sickness may have in common with several other forms of disease and injury, as far as the pathogenetic mechanisms as well as repair mechanisms are concerned (and where it is possible, at least in animal experiments and sometimes also in human patients, to achieve very much either for reducing the lethality, as I could observe myself when I was in Africa, or for reducing the severity of permanent organ damage), rather than on the assumed uniqueness of radiation sickness as a foe believed to be so strong that it can not be defeated.

It must be expected, however, that when a patient (or an experimental animal) is taurine-depleted as a direct consequence of exposure to high doses of ionizing radiation, all the anti-inflammatory effects of taurine that have been discussed will be correspondingly weakened, with the taurine depletion itself leading to an enhancement of the strength of the post-injury inflammatory response, leading also to enhancement of the risk that the patient may die not from the direct effects of ionizing radiation per se, but because of severe inflammation leading in turn to severe organ dysfunction, e.g. in form of diarrhoea.

Functions of taurine as an osmolyte and as a growth factor that may limit leukocyte growth when the supply is inadequate

One of the earliest studied functions of taurine Citation587 was its role as an osmolyte important not only for its contribution to the total osmolality of animal cells, but also for cellular volume regulation Citation587 Citation666 Citation667 Citation668 . Taurine has later been reported to function as a growth factor for leukocytes or leukocyte progenitor cells Citation669 Citation670 Citation671 Citation672 Citation673 and apparently also for other cell types including nerve and nerve progenitor cells Citation674 Citation675 Citation676 , with this growth stimulating effect having been observed even under adverse conditions with hypoxia and reoxygenation Citation677.

It is not unreasonable that these two apparently completely different functions of taurine might be mechanistically related because of the way by which cellular osmolyte content and volume now has been reported to regulate processes of cellular protein synthesis and apoptosis, at least in liver cells Citation678 Citation679 Citation680 Citation681 . The nature of these phenomena, even though it may be possible that they have been well studied only in hepatocytes, suggest that one might be dealing with very fundamental regulatory mechanisms, which presumably also could be very phylogenetically ancient. It is therefore reasonable to believe that they might be found also in all other mammalian nucleated cell types.

It has been found that when alterations of hepatocyte volume are induced either by anisoosmotic environments or under the influence of hormones, concentrative amino acid uptake and oxidative stress function as independent signals which contribute to the regulation of liver cell function and gene expression Citation681. Several long-known but mechanistically poorly understood effects of amino acids, which could not be related to their metabolism, such as the stimulation of glycogen synthesis or the inhibition of proteolysis, can now be explained as due to their effects on hepatocyte hydration, because they are quantitatively mimicked by swelling the cells in hypoosmotic media to the extent as the amino acids do Citation681. Likewise, transmembrane ion movements under the influence of hormones have been found to be an integral part of hormonal signal transduction mechanisms with alterations of cellular hydration acting as some kind of "second messenger" of hormone action Citation681. Integrins act as osmosensors for hepatocyte swelling and trigger activation of mitogen- activated protein kinase systems that function as osmosignaling cascades towards choleresis and autophagy inhibition Citation681. The opposite process, viz. hepatocyte shrinkage, triggers endosomal acidification as a signal for a ceramide-dependent activation of NADPH oxidase isoenzymes, which results in an oxidative stress signal with proapoptotic effects Citation681. Disturbances of osmosignaling and osmosensing are involved in a variety of pathophysiological conditions such as insulin resistance, protein catabolic states and cholestatic liver injury Citation681.

It should be added that protein degradation has been found to be heavily redox-regulated in other cell types, such as skeletal muscle cells, with the rate of intracellular protein degradation being enhanced by oxidative stress. When ROS production is enhanced because of NADPH oxidase activation following hepatocyte shrinkage, it is therefore reasonable to expect that this will not only enhance the likelihood that the cell will undergo apoptosis, but also enhance the rate of cellular protein degradation, even in the absence of apoptosis.

There are two different types of protein-degrading multiprotein complexes called proteasomes, viz. the 26S and 20S proteasomes. They depend on completely different forms of labelling for finding their target molecules. Protein molecules are targeted for degradation by 26S proteasomes by attachment of a protein called ubiquitin. The synthesis of some of the proteins needed for protein degradation by 26S proteasomes or otherwise is enhanced by the transcription factor NF-kappaB Citation682 Citation683 Citation684 , which can itself be activated by oxidative stress Citation685 Citation686 Citation687 . Enhanced oxidative stress wil therefore lead to enhanced degradation of muscle protein. NF-kappaB activation is involved also in the mechanism of disuse atrophy in the muscle Citation684 Citation688. Protein molecules are apparently targeted by oxidative damage for degradation by 20S proteasomes in different types of eukaryote organisms, including plants as well as animals Citation689 Citation690 Citation691 Citation692 , while the expression of other autophagy-related genes is enhanced by the oxidatively regulated protein kinase p38alpha/beta MAPK Citation693.

The combination of these mechanisms can explain why oxidative stress is a primary trigger Citation693 of cachectic muscle wasting, but the same signal pathways would be expected to control the rate of protein degradation in other cell types as well. Since it is common that disease situations leading to cachexia not only will be associated with enhanced degradation of protein as such, but also more specifically with enhanced catabolism of sulphur amino acid to form sulphuric acid, there are good reasons to expect that cachexia development easily will be a self-accelerating process once it has started, unless the diet is good enough (with enough sulphur amino acids) to compensate for the enhanced oxidative degradation of sulphur amino acids. One possible reason for this could also be impaired synthesis of proteins that are part of the mitochondrial respiratory chain as a direct consequence of protein malnutrition Citation533, which will in turn lead to enhanced mitochondrial ROS production because of enhanced Ohmian resistance to the passage of electrons through the respiratory chain Citation533.

Since taurine is one of the most important organic osmolytes in many cell types (but not universally so), it may be expected that an elevation of the total taurine content of the cell because it helps the cell to expand also may help to stimulate the synthesis of proteins in this cell at the same time as protein degradation and apoptosis will be hindered. At the same time, it may be expected that taurine because of its antioxidant effect (as demonstrated by its antidote effect against several different toxic substances functioning as prooxidants, and also by its protective effect against tissue damage caused by ischemia and reperfusion) will help to counteract cell protein degradation both by 26S and 20S proteasomes, as well as by other autophagy mechanisms.

Again it must be expected that radiation-induced taurine depletion will be harmful because it may inhibit the recovery of organ systems and physiological functions depending on very fast cellular growth, especially in the the immune system and perhaps also in the intestinal mucosa.

Can the antifibrogenic effects of taurine be useful during treatment of cancer and COPD?

However, opposite effects of taurine on cell growth have also been reported; taurine can sometimes function as an inhibitor of cell growth Citation694 Citation695 Citation696 Citation697 , which may probably be explained by its role also as some kind of signal substance inhibiting the growth of certain cell types via receptors located either at the cell surface or inside, perhaps even including nuclear receptors. A more unspecific mechanism caused by its antioxidant effect (and leading to reduced activation of oxidatively regulated signal pathways involving proteins that function as positive regulators of cell growth) can, however, not be excluded, since several different cell surface receptor-associated proteins, protein kinases, protein phosphatases and transcription factors are known which are either directly or indirectly (via proteins that are upstream in the signal pathway) redox-regulated, and which are known to be of crucial importance either for cellular growth regulation or the regulation of apoptosis (so that stimulation of the signal pathways concerned will lead to enhancement of the rate of cell growth and/or suppression of cell death by apoptosis).

Many of these redox-regulated signal proteins (surface receptor-associated proteins, enzymes or transcription factors) are very important in tumour cells, which often seem to exploit them to optimize their own growth and survival by providing an intracellular oxidative or nitrative stress high enough to give good redox-regulated activation of growth-stimulating and antiapoptotic signal pathways, but not so much oxidative or nitrative stress that it directly will cause the death of the cell either through apoptosis or necrosis. Improving the antioxidant or antinitrative defense capacity of the tumour cells compared with this self-sought evolutionary optimum may then paradoxically lead to down-regulation of their growth rate and enhancement of their death rate because of apoptosis, especially when more than one parallel signal pathways can be simultaneously downregulated to obtain a synergistic (multiplicative) interaction, e.g. by simultaneous inhibition of several tyrosine kinases (which the present author has tried to exploit for experimental self-therapy, with very satisfactory results, for a disease with marked symptoms suggestive of cancer, but uncertain diagnosis since biopsies have been negative, using very high daily doses during treatment intervals of two days duration of an otherwise very non-toxic substance functioning as a non-specific tyrosine kinase inhibitor, viz. genistein). As examples of such directly or indirectly redox-regulated proteins may be mentioned ras proteins, several tyrosine kinases, tyrosine protein phosphatases (which in contrast to the corresponding tyrosine kinases are inhibited by oxidative stress), several isozymes of protein kinase C and the transcription factors NF-kappaB, AP-1 and Sp1. This topic is much too large, however, that a detailed discussion with literature references is possible here.

Since taurine has been reported to inhibit the growth of cell types such as fibroblasts Citation695 and vascular smooth muscle cells Citation694 Citation697, it might be speculated that taurine supplementation could be helpful in some of those diseases where fibrosis and/or smooth muscle hypertrophy are important parts of the disease process, as in chronic obstructive pulmonary disease (COPD), alcoholic liver disease and other poison-induced fibrosis (in the liver or elsewhere), arteriosclerosis (in which patient group other cardiovascular protective effects of taurine also might be highly relevant), fibrosis in parts of the skeletomuscular apparatus leading to impaired mobility (with fibrosis prevention potentially being a positive side effect if taurine is used regularly for treatment of muscular or joint pains), and fibrosis as a side effect of radiation therapy.

There are already several animal experimental studies, especially with bleomycin-induced Citation63 Citation64 Citation66 Citation68 Citation69 Citation70 Citation71 Citation72 Citation75 Citation76 Citation77 Citation78 and amiodarone-induced Citation88 lower airway fibrosis, but also with CCl4-induced Citation30 Citation141 Citation144 Citation147 Citation148 and alcohol-induced Citation30 Citation39 fibrosis in the liver, as well as with cisplatin-induced fibrosis in the kidneys Citation51, suggesting that taurine may be protective not only in those animal experiments where such an effect has been found, but most likely also in several human diseases attended by fibrosis. These studies suggest that taurine supplementation also might help to protect against radiation-induced fibrosis-which has indeed been demonstrated in animal experiments Citation9, but might be expected to happen in human patients as well, e.g. in cancer patients following radiation therapy.

An obviously relevant question here is how better taurine status might influence the radiation sensitivity of the tumour cells. It is not possible to find out without carrying out the appropriate experiments whether taurine supplementation in cancer patients undergoing radiation therapy really may help to improve the therapeutic ratio (between desired therapeutic effect and unwanted side effects) or not. But it is not a priori implausible that taurine supplementation might be found to be more protective for normal tissues than for the tumour cells. One reason for this is that even if the tumour cells should have good enough uptake capacity for taurine that dietary supplementation will lead to enhanced intracellular taurine concentration and improved antioxidant protection of the tumour cells during irradiation, it must also be expected that enhancement of the taurine concentration inside the tumour cells may lead to reduced activation (as a direct consequence of the irradiation) of oxidatively activated regulatory proteins with anti-apoptotic effect, such as NF-kappaB, which will enhance the chance for initiation of the apoptosis programme as a consequence of radiation-inflicted injury to the DNA molecules of the tumour cells (genistein supplementation can probably be used to obtain even more of this effect, similarly as experiments with tumour cells in vitro have shown is possible for enhancing tumour cell killing by cytotoxic drugs). And it is not impossible that the last effect paradoxically might be more important than the first one for the overall death rate among the tumour cells following radiation therapy.

A cause-and-effect relationship has been proposed between cellular oxidative damage and increased fibrogenesis based on the fact that experimental treatment with antioxidants either prevents or quenches the fibrotic process Citation698. With some peculiarities in the different organs, fibrosclerosis is essentially the result of the interaction of macrophages and extracellular matrix-producing cells Citation698. The cross-talk is mediated by fibrogenic cytokines, among which the most important appears to be transforming growth factor beta 1 (TGF-beta 1) Citation698. When different types of macrophages are treated with 4-hydroxynonenal, which is a major aldehyde end product of membrane lipid oxidation, this treatment has been found consistently to induce both mRNA expression and synthesis of TGF-beta 1 Citation698. It is therefore reasonable to expect that taurine also may have an antifibrogenic effect because it functions as an important intracellular antioxidant that helps to reduce the rate of lipid peroxidation and hence the rate of 4-hydroxynonenal production.

Selenium and other antioxidant nutrients would be expected to have similar antifibrogenic effects as taurine and interact synergistically with it because they also will help to reduce the rate of lipid peroxidation and 4-hydroxynonenal production, which will help to limit the expression of the fibrogenic cytokine TGF-beta, while a high total intake of polyunsaturated fatty acids (PUFAs) or a high dietary PUFA/oleic acid ratio, other factors being equal, can be expected to enhance the rate of 4-hydroxynonenal production and hence probably will be profibrogenic. It might be speculated that it is not only because of much smoking that there are many COPD patients in Norway, but also because of the average composition of the Norwegian diet with high total PUFA intake, high average PUFA/oleic acid dietary ratio and not especially high average selenium and taurine intakes. It would be interesting to test this hypothesis epidemiologically by comparing the incidence of COPD among smokers (as a function of the total cumulate number of cigarettes) in Norway and Japan.

It is possible that all this may be very relevant for treatment of chronic obstructive pulmonary disease (COPD), if one wants to limit the rate of progression of this disease following early diagnosis. An ideal diet for COPD patients should presumably contain much taurine and much selenium at the same time as the dietary PUFA/oleic acid ratio should be much lower than is common in Norway and several other industrial countries today in order to minimize rates of 4-hydroxynonenal and TGF-beta 1 production. For COPD patients, it is not unreasonable that the antiischemic protective effect of taurine Citation209 Citation210 Citation211 Citation212 Citation213 Citation214 Citation215 Citation216 Citation217 Citation218 Citation219 Citation220 Citation221 Citation222 Citation223 Citation224 Citation225 Citation226 Citation227 Citation228 Citation229 Citation230 Citation231 Citation232 Citation233 Citation234 Citation235 Citation236 Citation237 Citation238 Citation239 Citation240 Citation241 Citation242 Citation243 Citation244 Citation245 Citation246 Citation247 Citation248 Citation249 Citation250 Citation251 Citation252 Citation253 Citation254 Citation255 Citation256 Citation257 Citation258 Citation259 Citation260 Citation261 Citation262 Citation263 Citation264 Citation265 also might be highly relevant, if taurine supplementation could help different organs to function better under conditions of tissue hypoxia developing as a consequence of impaired lung function. A cocktail that in addition to taurine also includes other antiischemic protectants such as selenium Citation272 Citation276 Citation277 Citation278 Citation279 Citation280 Citation281 Citation282 Citation283 Citation284 Citation285 Citation286 Citation287 Citation288 Citation289 Citation290 Citation291 Citation292 Citation293 Citation294 Citation295 Citation296 Citation297 Citation298 Citation299 Citation300 Citation301 Citation302 Citation303 Citation304 Citation305 Citation306 Citation307 Citation308 Citation309 Citation310 Citation311 and melatonin Citation346 Citation348 Citation349 Citation350 Citation351 Citation352 Citation353 Citation354 Citation355 Citation356 Citation357 Citation358 Citation359 Citation360 Citation361 Citation362 Citation363 Citation364 Citation365 Citation366 Citation367 Citation368 Citation369 Citation370 Citation371 Citation372 Citation373 Citation374 Citation375 Citation376 Citation377 Citation378 Citation379 Citation380 Citation381 Citation382 Citation383 Citation384 Citation385 Citation386 Citation387 Citation388 Citation389 Citation390 Citation391 Citation392 Citation393 Citation394 Citation395 Citation396 Citation397 Citation398 Citation399 Citation400 Citation401 Citation402 Citation403 Citation404 Citation405 Citation406 Citation407 Citation408 Citation409 Citation410 Citation411 Citation412 Citation413 Citation414 Citation415 Citation416 Citation417 Citation418 Citation419 Citation420 Citation421 Citation422 Citation423 Citation424 Citation425 Citation426 Citation427 Citation428 Citation429 Citation430 Citation431 Citation432 Citation433 Citation434 Citation435 Citation436 Citation437 Citation438 Citation439 Citation440 Citation441 Citation442 Citation443 Citation444 Citation445 Citation446 Citation447 Citation448 Citation449 Citation450 Citation451 Citation452 Citation453 Citation454 Citation455 Citation456 Citation457 Citation458 Citation459 Citation460 Citation461 Citation462 Citation463 Citation464 Citation465 Citation466 Citation467 Citation468 Citation469 Citation470 Citation471 Citation472 Citation473 Citation474 might possibly be even better for the COPD patient.

At the same time, COPD patients should probably also have a diet rich in alkaline-ash foods in order to compensate both for respiratory acidosis and metabolic acidosis caused by enhanced lactic acid production. This might presumably help to protect them against osteoporosis and reduce the overall level of vanilloid receptor-mediated C-fibre activation, leading in turn to reduction of the reflex-mediated secretion of glucocorticoids that is initiated by C-fibre stimulation Citation533. With reduction of the total level of C-fibre activation because of higher average extracellular pH, it must, moreover, be expected that there will be less ergoreflex-mediated adrenergic stimulation of the heart leading to reduction of tachycardia, and also less subjective experience of respiratory distress.

Since the sensitivity of C-fibres to stimulation via the vanilloid receptor is enhanced by prostaglandins and by activation of protein kinase C (PKC) inside the C-fibres Citation533 Citation534, it will probably also be useful for COPD patients if the diet can be modified so that the rate of prostaglandin production and the degree of oxidative stress-mediated or diacylglycerol-induced activation of PKC inside the C-fibres also can be minimized – for reducing glucocorticoid production, tachycardia and sensation of respiratory distress. This will probably be helpful also for enhancing the subjectively experienced training tolerance of the patient, making it easier to use resistance training for prophylaxis and treatment of COPD-associated cachexia.

To obtain this, the dietary ratio arachidonic acid/(long-chain omega-3 PUFAs) should be strongly reduced compared to what is common in several Western countries today, not only by enhancing the intake of long-chain omega-3 PUFAs, but also reducing the intake of arachidonic acid at the same time as the intake of antioxidant nutrients including selenium should be high Citation534. The blood sugar level should be kept down at a moderate level both for reducing lactic acid production in poorly oxygenated tissues (which will directly lead to more activation of the vanilloid receptor) Citation534 and because it is possible that it may contribute to enhanced production of diacylglycerol in the C-fibres and hence enhancement of the sensitivity of the C-fibres to vanilloid receptor-mediated stimulation Citation534.

Safety of taurine preparates

Taurine has been used so much in animal experiments and also as part of the culture medium used for studies on cells in vitro (e.g. cardiomyocytes) that experimental scientists have gained much experience with the toxicity properties of this substance in non-human animal species and cell cultures. The experience from such studies is that the toxicity of taurine must be very low both for non-human animal species and isolated cells grown in vitro.

The safety of taurine for humans can be estimated partly on basis of knowledge about the composition of the diets of hunter and gatherers before the agricultural and industrial revolutions and about the health of such populations, partly from epidemiological studies on the statistical association between taurine intake and health in several different countries today, and partly on basis of reports about possible adverse side effects among people who have ingested high doses of taurine (much above normal intakes) from dietary supplement preparates or “energy drinks” rich in taurine.

There can be no doubt that the taurine intake of hunters and gatherers living on so-called Paleolithic diets Citation699 Citation700 Citation701 Citation702 Citation703 Citation704 typically must have been very high, especially for people who have been living in areas with Sub-Arctic or Arctic climate where animal foods were more easily available as food energy sources compared to carbohydrate-rich or fat-rich plant foods such as nuts, root tubers, legume seeds and cereals. It has been estimated that about 2/3 of the food energy intake of hunter-gatherers living in the Kalahari desert typically came from plant foods and 1/3 from animal foods Citation705, but for Inuits both at the coast and in the inland (Inuit reindeer hunters in Alaska), it must probably have been opposite with at least 2/3 of the food energy intake or more coming from animal foods.

However, it has been common that Sub-Arctic and Arctic hunter-gatherers have been eating much of other plant foods, whatever has been available (including the content of grouse intestines among Inuits in Greenland) Citation706 Citation707, which even though they were not very important as sources of food energy still may have been important for the dietary intake not only of vitamin C, folate and polyphenolic plant antioxidants, but also of some mineral nutrients, such as manganese, nickel and copper, that are generally more abundant in plant foods than in animal foods Citation623, with manganese and nickel being the most extreme examples Citation623 – with more than 90% of the dietary intake of manganese in Finland during the 1970s coming from plant foods, spices and plant-derived beverages such as coffee and tea Citation623. It is reported that a group of Inuits living under especially harsh climatic conditions far north in Arctic Canada even were eating grouse faeces Citation707, which conceivably might have been important to prevent manganese deficiency in a place where little plant foods were available because of the exceptionally harsh climate.

For Sub-Arctic and Arctic hunters, it is reasonable to assume that the daily taurine intake commonly might have been around 1 g per day or more. The general health status of Inuits has nevertheless been good, especially when considering the risk of cardiovascular disease Citation708 Citation709, and there is nothing to suggest that their high intake of taurine might have been harmful for their health. Nor is there any suggestion that other “Paleolithic diets” rich in meat, offal and/or seafood and hence rich in taurine have been associated with health problems that might be a consequence of adverse consequences of high taurine intake Citation699. But there is reason to believe that vitamin D deficiency may have been a severe threat to several populations living in the inland in cold parts of Eurasia during the Ice Age.

In the CARDIAC (CVD and Alimentary Comparison) Study, which was a WHO-coordinated multi-center epidemiological survey on diets and cardiovascular risks and mortalities in 61 populations from different parts of the world, it was found that twenty-four-hour urinary taurine excretion (24U-T) was significantly inversely related with coronary heart disease mortality Citation572. Higher 24U-T excreters had significantly lower body mass index, systolic and diastolic blood pressure, total cholesterol, and atherogenic index (total cholesterol/high density lipoprotein-cholesterol) than lower taurine excreters Citation572. Protective effects of taurine on cardiovascular disease risks were found to be intensified in individuals whose 24U-T and -magnesium excretions were higher. One of the groups studied (which was a group of Australian aboriginals with high consumption of seafood) was found to have an average taurine intake during a 2 week study period as much as about 3200 mg per day, far higher than the average for CARDIAC study populations in the rest of the world Citation572.

It can be concluded from this large study that there is no indication of any health hazard associated even with the highest taurine intake encountered in the study populations (up to as much as 3.2 g taurine per day).

There are, however, some reports about adverse events following consumption of large quantities of taurine-rich so-called energy drinks. It is far from certain that this happened because of high taurine ingestion. There is a possibility that sudden death happened for causes unrelated to energy drink consumption, that it might be related to specific situations in which energy drinks are used, such as ingestion of the energy drink in combination with alcohol Citation710 or perhaps illegal drug abuse, and also that it may have been caused by too high intake of other substances also found in substantial amounts in the energy drink, such as caffeine.

In a recent review article about risks associated with consumption of energy drinks in youths, it is concluded that although the issue of taurine-induced toxic encephalopathy has been addressed, it is likely that the risk of taurine toxicity after energy drink consumption remains low Citation710. However, whether the prolonged use of energy drinks providing more than 3g taurine daily is safe remains to be examined in the future Citation710. The consumption of energy drinks may increase the risk for caffeine overdose and toxicity in children and teenagers Citation710. The practice of consuming great amounts of energy drink with alcohol is considered by many teenagers and students a primary locus to socialize and to meet people Citation710. This pattern of energy drink consumption explains the enhanced risk of both caffeine and alcohol toxicity in youths Citation710. 25 to 40% of young people report consumption of energy drink together with alcohol while partying Citation710. Consumption of energy drinks with alcohol during heavy episodic drinking is associated with enhanced risk of serious injury, sexual assault, drunk driving, and death Citation710. However, even after adjusting for alcohol consumption, students who consume alcohol mixed with energy drinks had dramatically higher rates of serious alcohol-related consequences Citation710. It has been reported that the subjective perceptions of some symptoms of alcohol intoxication are less intense after the combined ingestion of the alcohol plus energy drink; however, these effects are not detected in objective measures of motor coordination and visual reaction time Citation710. The possibility should perhaps not be excluded that caffeine and taurine in combination might lead to reduction of subjectively experienced symptoms of alcohol intoxication sufficiently to enhance the risk of serious alcohol overdose, leading in turn to serious acute disease or death.

When taking into consideration all the observations mentioned above both from experimental studies, epidemiological studies of large human populations and reports about possible adverse side effects of energy drink consumption, it may be concluded that the safety of taurine preparates is very good, and better-as regards the risk of serious side effects-than for a vast majority of commonly used non-nutrient drugs. This is especially important when comparing the potential hazards attending the use of high-dose taurine preparates with such ordinary drugs that are known to have mutagenic effects, such as those cytotoxic drugs that are currently used not only for treatment of otherwise lethal cancer, but also for treatment of the non-lethal disease rheumatoid arthritis, and paracetamol Citation534, which is regularly consumed at high daily doses, often without prescription, by a huge number of patients all over the world – whereas taurine very likely is antimutagenic for reasons that have been explained in detail above. However, very high daily doses of taurine should not be combined with too much alcohol, too much caffeine or illegal recreational drugs.

It is also well known that several other nutrients can have toxic effects when ingested at dosage levels much higher than a normal dietary intake, and one can not exclude the possibility that this might also be the case with taurine, or that there might be paradoxical interaction effects leading to enhanced toxicity of other substances ingested in combination with taurine. Such hypothetical and unknown hazards should, however, be weighted against the known risks or certainly known harmful consequences of the disease or injury where high-dose taurine supplementation might be considered to be used as part of the therapy, and against the possible therapeutic benefits obtained by using taurine.

With something as serious as the health damage caused by excessive levels of ionizing radiation, there can be little doubt what the answer to this question should be (and the same would of course also be the case with life-threatening conditions, such as very serious burn injury, life-threatening asthma, malignant avian influenza or SARS).

There is, however, a potential risk that a sudden large enhancement of taurine intake might have adverse effects in patients suffering from insulin-dependent diabetes. This is because taurine has been reported to bind to the insulin receptor Citation661 and affect blood sugar regulation, at least in experimental animals Citation659 Citation660; there is therefore a possibility more than purely theoretical that administration of high doses of taurine to an insulin-dependent diabetic unfamiliar with the substance might provoke unexpected and potentially dangerous hypoglycemia. The best way to avoid this hazard is probably to reduce the insulin dosage temporarily before starting taurine therapy and subsequently readjust it as required, while keeping the patient under good surveillance and informing him about the possible risk, so that he may modify his carbohydrate intake as needed if he notices symptoms of incipient hypoglycaemia.

Administration of taurine preparates

Administration of taurine to patients suffering from severe H5N1 pneumonia, SARS or asthma might be done either perorally, by inhalation of an aerosol spray preparate or parenterally. Inhalation preparates have been tried in asthma with good results Citation586 and would be expected to give an even more rapid effect in the bronchi and lungs compared to peroral administration. But considering the pharmacokinetic behaviour of taurine and its low toxicity, there seems to be no other good reason to prefer inhalation taurine preparates (when considering taurine itself, as different from taurine derivatives that might be degraded in the gastrointestinal tract or liver, and that might be given to the patients for other reasons than because of the effect of taurine itself) rather than peroral or parenteral administration.

For patients suffering from diarrhoea, it should be possible to give them taurine either perorally or by parenteral administration. For conscious patients with a normally functioning intestine, it must certainly be most practical to give this substance by the oral route either alone or in combination with other nutrients or drugs. For unconscious or sleeping patients, parenteral administration should probably be preferred when practically feasible, and the same might also be the case when the intestine is severely damaged in a way leading to malabsorption (even though it is possible that the local effect of high taurine concentration in the intestinal lumen might be useful in some of these patients).

Given the low toxicity of taurine, its numerous protective functions and the facility with which taurine deficiency can develop because of enhanced leakage from individual cells and enhanced urinary excretion in various disease situations or because of therapy (especially during treatment of cancer patients with radiation therapy or cytotoxic drugs), it would probably be right to add taurine as a matter of routine to preparates for parenteral nutrition, as well as to food preparates to be given through nasogastric tube. The reported antidiarrhoeal effect of taurine Citation662, which is consistent with the very rapid recovery of the patients observed by the present author when using a diet rich in taurine in combination with oral rehydration solution for treatment of a large group of children suffering from the combination of malnutrition, measles and diarrhoea in Africa Citation663, needs further testing in animal models as well as in human patients. But if this effect can be confirmed, it may be possible that it could also be therapeutically useful to add taurine to preparates used for oral rehydration of cholera or other diarrhoea patients.

The pharmacokinetics of this substance with normally good intestinal absorption, but rapid urinary excretion (when it is given at a high dosage level) should be kept in mind; for oral administration of taurine preparates (being perhaps not equally important when abundant taurine is given as part of a food mixture with enough bulk that it will not pass too rapidly through the stomach into the intestine), it might therefore be useful to have a slow-release formulation securing that there will be a high plasma taurine concentration for a more extended period of time. Such formulations are perhaps not commercially available in any country in the world today, but WHO and the health authorities of individual countries should encourage pharmaceutical companies to make them. The dosage level for severely ill adult patients should probably not be less than about 1 g taurine per day. Taurine appears to be exceptionally non-toxic, which means that the risk of harmful side effects may be judged very small, except possibly for insulin-dependent diabetics, for reasons which earlier have been explained.

Taurine should not be used alone, but as part of a multifactorial strategy for treatment of severe radiation injury

In all of its known physiological functions, taurine collaborates with several other nutrients and/or hormones. Several other dietary deficiencies can therefore mimick taurine deficiency in some of their functional consequences, and the antioxidative protective hormone melatonin can mimick some of the positive therapeutic effects of taurine supplementation, not only for protection against tissue injury caused by ischemia/reperfusion Citation346 Citation348 Citation349 Citation350 Citation351 Citation352 Citation353 Citation354 Citation355 Citation356 Citation357 Citation358 Citation359 Citation360 Citation361 Citation362 Citation363 Citation364 Citation365 Citation366 Citation367 Citation368 Citation369 Citation370 Citation371 Citation372 Citation373 Citation374 Citation375 Citation376 Citation377 Citation378 Citation379 Citation380 Citation381 Citation382 Citation383 Citation384 Citation385 Citation386 Citation387 Citation388 Citation389 Citation390 Citation391 Citation392 Citation393 Citation394 Citation395 Citation396 Citation397 Citation398 Citation399 Citation400 Citation401 Citation402 Citation403 Citation404 Citation405 Citation406 Citation407 Citation408 Citation409 Citation410 Citation411 Citation412 Citation413 Citation414 Citation415 Citation416 Citation417 Citation418 Citation419 Citation420 Citation421 Citation422 Citation423 Citation424 Citation425 Citation426 Citation427 Citation428 Citation429 Citation430 Citation431 Citation432 Citation433 Citation434 Citation435 Citation436 Citation437 Citation438 Citation439 Citation440 Citation441 Citation442 Citation443 Citation444 Citation445 Citation446 Citation447 Citation448 Citation449 Citation450 Citation451 Citation452 Citation453 Citation454 Citation455 Citation456 Citation457 Citation458 Citation459 Citation460 Citation461 Citation462 Citation463 Citation464 Citation465 Citation466 Citation467 Citation468 Citation469 Citation470 Citation471 Citation472 Citation473 Citation474 , but also for protection against ionizing radiation Citation711 Citation712 Citation713 Citation714 Citation715 Citation716 Citation717 Citation718 Citation719 Citation720 Citation721 Citation722 Citation723 Citation724 Citation725 Citation726 Citation727 Citation728 Citation729 Citation730 Citation731 Citation732 Citation733 Citation734 Citation735 Citation736 Citation737 Citation738 Citation739 Citation740 Citation741 Citation742 Citation743 Citation744 Citation745 Citation746 Citation747 Citation748 Citation749 Citation750 Citation751 Citation752 Citation753 Citation754 Citation755 Citation756 Citation757 Citation758 Citation759 Citation760 Citation761 Citation762 Citation763 Citation764 Citation765 Citation766 Citation767 Citation768 Citation769 Citation770 Citation771 . For obtaining an optimal therapeutic response following severe radiation injury, it will not be helpful to think of taurine as some kind of “magic bullet” that can be used alone, in spite of numerous different beneficial effects. Rather, one has to combine everything that is needed for optimizing some particular physiological function (or even for allowing it to occur).

Protective effects during or following exposure to ionizing radiation have been reported also for selenium Citation772 Citation773 Citation774 Citation775 Citation776 Citation777 Citation778 Citation779 Citation780 Citation781 Citation782 Citation783 Citation784 Citation785 Citation786 Citation787 Citation788 Citation789 Citation790 Citation791 Citation792 Citation793 Citation794 , glutathione and glutathione precursors Citation795 Citation796 Citation797 Citation798 Citation799 Citation800 Citation801 Citation802 Citation803 Citation804 Citation805 Citation806 and carnosine Citation807 Citation808, which are substances that earlier have been mentioned as examples of antioxidative nutrients that offer significant protection against tissue damage caused by ischemia and reperfusion, similarly as has been found for taurine in a large number of experimental studies. However, not all reported observations are consistent regarding radioprotective effects of glutathione (which apparently depend on experimental conditions and are not always observed). Carnitine and carnitine derivatives Citation809 Citation810 Citation811 Citation812 Citation813 Citation814 Citation815 Citation816 , and numerous plant antioxidants (literature so abundant that references are omitted here, but it can easily be found on PubMed) have also been reported to function as radioprotectants.

The radioprotective effects of antioxidant nutrients and the antioxidant hormone melatonin can in large measure be explained by their effect on the cellular capacity to scavenge harmful reactive species that are formed because of the passage of ionizing radiation through cells Citation817, such as hydroxyl radical (OH.), peroxyl radical (HO2 .), superoxide anion radical (O2 -.). hydrogen peroxide (H2O2), singlet oxygen and peroxynitrite Citation475. Some of these radioprotective substances, such as selenium and glutathione, act mainly via antioxidant scavenger enzymes, such as glutathione peroxidases and peroxiredoxins Citation817. Carnosine Citation818 Citation819 Citation820 Citation821 Citation822 Citation823 Citation824 Citation825 , melatonin Citation713 Citation714 Citation715 Citation719 Citation720 Citation721 Citation727 and numerous plant-derived antioxidants Citation826 function themselves as scavengers of some of those harmful molecules that are produced either more directly by ionizing radiation (such as hydroxyl radical and peroxyl radical) or by radiation-induced lipid peroxidation, including organic free radicals and in the case of carnosine such reactive carbonyl compounds that can be formed as secondary products of lipid peroxidation Citation523, with carnosine Citation818 Citation819 Citation820 and melatonin Citation713 Citation714 Citation715 Citation719 Citation720 Citation721 Citation727 both being good scavengers of hydroxyl radical and peroxyl radical.

Melatonin has, moreover, also been reported to scavenge superoxide anion radical, singlet oxygen, and peroxynitrite Citation720, and carnosine is reported to function as a scavenger of peroxynitrite Citation827, which is formed when peroxyl radical or superoxide anion radical produced by radiation reacts with NO that has been produced by NO synthases in radiation-exposed cells or some of their neighbours (because NO is very freely diffusible and can diffuse over some distance from one cell to another before it is scavenged). Only poor scavenging of peroxynitrite by carnosine was, however, found in another study, where, nevertheless, carnosine was found to be a good scavenger of hypochlorite Citation828, which may not be relevant for radiation injury, but could be so for non-infectious inflammatory diseases, such as rheumatoid arthritis and asthma. Carnosine has, moreover, also been reported to function (similarly as glutathione also does) as a scavenger of NO Citation829 Citation830, which will help to reduce the rate of peroxynitrite production, while melatonin reduces the rate of NO formation by reducing the activity of neuronal Citation831 Citation832 Citation833 Citation834 and endothelial Citation835 NO synthase. This happens apparently because melatonin binds to calmodulin Citation832 Citation833 Citation836 and hence either modifies the stability of the activated (NO-producing) complex between a calcium-calmodulin complex (containing 4 Ca+ +  ions) and the enzyme itself (which possibly could mean that a higher calcium concentration might be needed for activation of the enzyme) or changes the conformation of this active enzyme complex in a way leading to reduction of the activity of the calcium/calmodulin-activated enzyme (in which case the calcium concentration needed for activation might remain the same, while the effect of calcium influx into the cell on NO production nevertheless will be reduced because the kinetic properties of the active enzyme have been changed). Melatonin exerts a similar regulatory effect also on at least one of those other enzymes that are activated by the calcium-calmodulin complex, viz. Ca2 + /calmodulin-dependent protein kinase II Citation837. Peroxynitrite reacts with DNA molecules without any need for assistance from iron or other redox-active transition metals that function as catalysts of DNA oxidation by H2O2. It is a powerful mutagen which oxidizes DNA bases and can produce both single- and double-stranded breaks in the DNA molecule Citation838 Citation839 Citation840 Citation841 Citation842 Citation843 .

Yet another class of radioprotectant molecules (probably including taurine and the histidine-containing dipeptides carnosine and anserine) may work by counteracting the prooxidant catalytic effect of iron or other redox-active transition metals. But carnosine and taurine have a double mechanism of action for their radioprotectant effects since they also function as scavengers of some of those reactive species that can react with DNA molecules or enhance the rate of lipid peroxidation.

Melatonin, when acting as a hormone via nuclear receptors, helps, moreover, also to enhance the expression, at least in some cell types and organs (such as the brain), of several enzymes that either function as antioxidant scavengers themselves (such as superoxide dismutase and glutathione peroxidase) or participate in the electron transport chain needed for the function of some of the scavenger enzymes (such as glutathione reductase and glucose-6-phosphate dehydrogenase, which in most human cells are needed for normal regeneration of GSH from GSSG in the cytosol and hence are required for the function of glutathione peroxidase in the cytosol) Citation715 Citation719.

It must be expected from this that there will be a number of synergistic (multiplicative) interactions between various antioxidant nutrients and melatonin when functioning as radioprotectants. Selenium and glutathione will interact synergistically because of the tert-uni ping pong kinetics of glutathione peroxidases Citation534, that scavenge H2O2, organic hydroperoxides and peroxynitrite Citation534, and melatonin will interact synergistically with these antioxidant nutrients when it enhances the expression, at least in some organs, of glutathione peroxidase, glutathione reductase and glucose-6-phosphate dehydrogenase.

For peroxynitrite there will also be two other effects of melatonin interacting synergistically with the above-mentioned one: when it reduces NO production by neuronal and endothelial NO synthase and when it functions itself as a scavenger of peroxynitrite. When melatonin has multiple different effects helping to reduce the concentration of peroxynitrite in cells expressing neuronal NO synthase, it means that the dose-response curve for peroxynitrite concentration in brain cells as a function of melatonin concentration in blood plasma will be strongly nonlinear. There is reason to believe that this kind of non-linear dose-response curve (which will help this hormone to function much like an on-off switch) may be very important for understanding the effects of melatonin as a regulator of diurnal biological rhythms.

The non-linear off/on switch-like shape of this curve suggests also that the concentration of peroxynitrite in brain cells may be one of the most important targets of melatonin when acting as a regulator of diurnal rhythms. One possible explanation might be that peroxynitrite (which will be produced abundantly in brain nerve cells during the day following activation of NMDA receptors) could be critically involved in learning processes as one of the most important signal substances that start a chain of processes that will initiate subsequent protein synthesis needed for those microstructural anatomical changes (with the synaptic contacts between some pairs of nerve cells being improved because of enhancement of the total number of synapses between the two nerve cells concerned) that are needed for establishment of long-term memory. But since peroxynitrite has also a number of harmful effects to the brain cells, a prolonged period with little peroxynitrite during the night might be needed for repair of those peroxynitrite-induced lesions that have accumulated in the brain during the preceding period of light, wakeful sensation and learning the day before.

Glutathione will also help to reduce the rate of peroxynitrite production because of its reaction with NO leading to S-nitrosoglutathione production, with S-nitrosoglutathione not forming peroxynitrite when it subsequently reacts with superoxide anion radical Citation520, and taurine will probably interact synergistically with H2O2-scavenging enzymes when protecting against the catalytic effect of iron against reactions between H2O2 and organic molecules.

The theoretical rationale for using a cocktail of different antioxidant nutrients and melatonin for obtaining optimal radioprotection rather than using high doses of only one of these substances alone would thus appear very strong. It is possible that this could have important practical implications not only for radioprotection following severe nuclear accidents, but also for reducing the side effects from radiotherapy in cancer patients – especially if this principle could be used for protecting normal tissues at the same time as other methods (e.g. inhibition of polyamine synthesis in the tumour cells while supplementing the patient with enough polyamines to maintain normal levels in the normal tissues Citation817), are used for selectively enhancing the radiosensitivity of the tumour cells.

Another good reason for preferring multifactorial treatment strategies to monofactorial therapeutic intervention (say, with high doses of one drug) is also that a good multifactorial treatment strategy often will be associated with much lower risk of adverse side effects than when high doses of only one pharmacologically active substance are used. This is because the different substances that are used as components for multiple intervention very commonly will have different side effects, even if they collaborate in synergistic fashion to produce the same desired therapeutic effect, and sometimes it is even possible that some of the substances that are used for the multiple intervention can protect against the side effects of other substances that are also used as part of the same multiple treatment strategy Citation534. When different substances interact synergistically with each other to produce the same therapeutic effect, it is possible to reduce the dosage level for each one of them with corresponding reduction of the risk of adverse side effects, and still obtain the same desired overall therapeutic response (or even a better one). Moreover, it is also common that nutrients, when used as part of multiple treatment strategies, are less toxic and therefore associated with less risk of adverse side effects (even when given at doses higher than normal dietary intakes), compared with most ordinary drugs. The same is also the case with the antioxidant and immunostimulating hormone melatonin and some of those other substances that can be used for immunostimulation.

From a theoretical standpoint there must therefore be very good reason to hope, for several different diseases and severe injury conditions, that well-designed multifactorial treatment strategies incorporating optimal nutrition as an important part of it might be able to improve the therapeutic ratio very substantially (with significant improvement of the therapeutic outcome at the same time as adverse side effects can be reduced) compared with such monofactorial or oligofactorial treatment methods that are used for the same diseases and injuries today Citation534. And they should in a majority of cases not need to be overly expensive (since some of those substances that often might be used can be produced very cheaply and are not protected by patents), which means that they also as regards their health economic cost-benefit ratio might be very favourable, making it possible for a society with limited economic and skilled manpower resources to take care of a larger number of patients than today at the same total cost, and yet achieve better average therapeutic results than now. While this is obviously even more important in poor countries than in the more affluent ones, the problem of resource constraints compared with the number of patients needing medical treatment and care is in principle the same almost everywhere, even in rich countries such as the United States and Norway, with only some very few possible exceptions.

There are two different (each of them very strong, but even more compelling when seen in combination with each other) reasons for confidence that the results of experiments for studying the radioprotective effect of individual substances mentioned above in other mammalian species can safely be extrapolated to humans. The first is that one is dealing with very fundamental physical, chemical and biochemical mechanisms that must be expected to be the same everywhere in all living cells and organisms that use the same set of antioxidative protective enzymes as we do. The other is the same as has been mentioned when discussing the evidence for protection by antioxidant nutrients and melatonin against permanent tissue damage caused by ischemia followed by reperfusion, viz. the combination both of the quality and abundance (i.e. number of independent studies) of animal experimental observations showing essentially the same when confirming the radioprotectant effect. The number of independent experimental studies is also in this case (as for antiischemic protection) especially large for melatonin.

It is possible that the inconsistent results of experimental studies of the radioprotective effect of glutathione partly or wholly might be explained by diverse effects of glutathione both as an antioxidant and a prooxidant, with not all effects being protective when a cell is exposed to ionizing radiation. Reduced glutathione (GSH) must be expected to be protective because of its function as reducing cofactor for H2O2-scavenging, organic hydroperoxide-scavenging and peroxynitrite-scavenging antioxidant enzymes such as glutathione peroxidases and peroxiredoxin-6 (1-Cys peroxiredoxin), and also by functioning as a complexing agent for ferrous iron (with the GSH/ferrous iron complex being, if not necessarily directly protective, at least far less active as a prooxidant compared to several other complexes between ferrous iron and small organic molecules, including the glutathione degradation products cysteinylglycine and cysteine). But GSH may also be expected to have an adverse effect, similarly as vitamin C does, by functioning as a reducing agent for ferric iron that is complex-bound to other organic molecules (although it is possible that the harm resulting from this effect may be far less for GSH than for ascorbate because of higher stability of the complex of ferrous iron with GSH than for the complex between ferrous iron and ascorbate).

However, it should in theory be possible to exploit the synergistic interactions between glutathione and other radioprotective substances to enhance its beneficial effects and reduce the possible harmful ones. If the levels of glutathione peroxidase and associated enzymes (such as glutathione reductase and glucose-6-phosphate dehydrogenase) can be enhanced by supplementation with selenium and/or melatonin, this would be expected to enhance the protective effect of glutathione when it helps to scavenge H2O2, organic hydroperoxides and peroxynitrite, and it is possible that taurine could be used for reducing its harmful effect when GSH functions as a reductant for ferric iron.

It may be noted that most species of seafish are good sources not only for taurine Citation550 but also for selenium Citation550 Citation551 Citation623, for glutathione precursor amino acids Citation550 Citation551, and when the fish has been freshly caught presumably also for glutathione itself. Histidine dipeptides, especially anserine, which has been very little studied but appears to have similar effects as carnosine as a radioprotectant Citation807, are found at very different concentration levels in differrent species of fish, but some species, e.g. capelin (Mallotus villosus), contain much anserine Citation844 Citation845 Citation846 Citation847 Citation848 Citation849 . Since sea fishes, in great contrast to humans and several other mammalian species living on land, do not normally eat much starch or sugar, it is common that they rely on protein and fat as their most important metabolic fuels. It should therefore be expected that pelagic fast-swimming species with good aerobic capacity must have large capacity for beta-oxidation of fatty acids, which means that they must contain much carnitine in order to get the fatty acids transported fast enough into their mitochondria (and probably even more so when the water temperature is low). Although it is difficult to find analytical data for carnitine concentrations in different types of fish and different fish organs, it may be expected that many fish species are excellent dietary sources of carnitine, with the concentration perhaps being especially high in pelagic species living in cold water, such as capelin, as well as in the fastest swimmers, such as tuna.

It may be concluded that the average intake of radioprotective nutrients probably may be higher in Japan than in most other countries in the world because of exceptionally high average seafood consumption, at the same time as the average intake of plant-derived antioxidants also is high in Japan (i.a. from soy products and tea). It is probable that this may have helped to reduce the health damage caused by high doses of ionizing radiation among survivors from Hiroshima and Nagasaki, and there is also good reason to expect that the composition of the normal diet in Japan is protective today for those who have been exposed to high levels of ionizing radiation while working with damaged nuclear reactors. But it is possible that this form of protection may be further improved by additional taurine supplementation, and there is in particular reason to expect that it might be improved by melatonin supplementation at a high dosage level (e.g. 20 or 40 mg melatonin per day for adult patients).

For people living in countries where seafood consumption is not as high as in Japan, FPC type B combined either with antioxidant-rich plant foods or plant antioxidants given as dietary supplements would probably be excellent for enhancing the total dietary intake of radioprotective antioxidative nutrients. The FPC dosage level for adult patients suffering from radiation injury should probably not be less than 100 g per day.

Nutrients needed for DNA replication and repair.

Normal cell growth cannot happen without an adequate supply of several different molecular building blocks, both macronutrients and micronutrients, and not only essential ones but also conditionally essential nutrients such as taurine and nucleotides. This is the same both when considering the growth of enterocyte progenitors and that of several different classes of leukocytes or their progenitor cells.

DNA repair is also a process dependent on several different nutrients (but apparently not, as far as is known, taurine). They are in large measure (since DNA repair cannot proceed normally without an adequate supply of monodeoxyribonucleotide building blocks) the same ones that are also required for DNA synthesis and therefore are needed for fast cellular growth, which in turn is absolutely required for fast recovery both of the immune system (if it has not been damaged beyond repair) and of the intestinal mucosa (again if it has not been damaged beyond repair). But DNA repair depends also on other enzymes that do not participate in DNA replication, and some of these other enzymes (that are needed for normal DNA repair) depend also on nutrient cofactors making them potentially vulnerable to malnutrition.

For minimizing the harmful biological effects of ionizing radiation by interventions carried out before, during or after exposure (e.g. before people shall go into a severely damaged nuclear power plant in an effort to try to hinder further escalation of the nuclear disaster), it will be helpful to try to do everything possible for optimizing antioxidant protection (not only during the exposure to radiation, but also afterwards in order to minimize tissue injury caused by the post-exposure inflammatory response) and also for DNA repair processes (both during and after the radiation exposure). After exposure to ionizing radiation has occurred, it will be important to continue to maintain an optimal supply of all nutrients that are needed for normal DNA repair. When much DNA damage occurs, this would be expected to lead to acutely enhanced demand for all mononucleotides that are needed for the DNA repair process, perhaps even so much as to induce mononucleotide deficiency that might limit the rate of normal DNA synthesis and cellular growth – which could represent a possible explanation both for immunosuppression and gastrointestinal damage following exposure to high doses of ionizing radiation.

The dietary requirement for nucleotides/nucleosides is therefore likely to be higher in patients who have suffered from severe radiation injury than in healthy persons, perhaps by a significant factor, at the same time as intestinal injury conceivably might be attended by malabsorption making it more difficult to absorb as much nucleotides or nucleosides that may be needed to allow both optimal DNA repair and optimal cell growth, e.g. in leukocytes and leukocyte progenitor cells. Intestinal malabsorption might, of course, also affect the absorption of nutrients needed for endogenous synthesis of deoxyribonucleotides by de novo pathways, such as glutathione precursor amino acids or glutathione itself (which participates in deoxyribonucleotide synthesis because it is needed as part of the electron transport chain from NADPH via glutaredoxin to ribonucleotide reductase), riboflavin (which is needed in both parallel pathways for electron transport from NADPH to ribonucleotide reductase either via glutaredoxin or thioredoxin), thiamine (which is needed for normal regeneration of NADPH from NADP+ because of the role of thiamine pyrophosphate as a cofactor in transketolase), folate (which is needed for conversion of uridylate to thymidylate) and vitamin B12 (which is also needed for conversion of uridylate to thymidylate).

If the cells contain too little deoxythymidylate when they shall synthesize or repair DNA, deoxyuridylate will be incorporated into the DNA molecule instead of deoxythymidylate, i.e. the DNA molecule will now contain uracil in what should normally have been a thymine position Citation850 Citation851 Citation852 Citation853 . This is a form of DNA damage that can be repaired, but the repair mechanism does not function perfectly, especially not when thymidylate continues to be deficient, and it may sometimes lead to DNA double-strand break instead of correct repair Citation850 Citation851 Citation852 . This can happen not only because of folate deficiency Citation850 Citation854, but also because of vitamin B12 deficiency Citation854 Citation855 Citation856 . A similar effect can, moreover, also be expected to occur because of zinc deficiency, since thymidine kinase, which is an enzyme needed for reutilization via the salvage pathway of thymidine for making DNA, is a zinc-dependent enzyme vulnerable to zinc deficiency Citation857.

It must be expected that there will be a synergistic interaction between zinc deficiency and low dietary intake of DNA as causes of reduced thymidylate synthesis by the salvage pathway, with the mutagenic effects of thymidylate depletion being especially severe when thymidylate synthesis both via the salvage and de novo pathways are simultaneously inhibited e.g. because of a combination both of zinc deficiency and vitamin B12 deficiency. This is something that easily can happen when the total dietary intake of animal foods is low Citation551 at the same time as the local agricultural soils are depleted in zinc because of the combined effects of prolonged chemical weathering under humid tropical conditions and deforestation Citation551 Citation858, which is a situation very commonly encountered in much of sub-Saharan Africa.

It can, moreover, be expected that there will be a synergistic interaction between those dietary deficiencies that specifically affect the synthesis of thymidylate and such dietary deficiencies that will cause reduction of the activity of ribonucleotide reductase and hence may lead to reduction of the synthesis of all deoxyribonucleotides needed for DNA synthesis and repair. In addition to those nutrients that are needed for normal electron transport to ribonucleotide reductase via glutaredoxin and already have been mentioned (glutathione and its precursor amino acids, riboflavin, niacin and thiamine), iron is also needed here because ribonucleotide reductase itself is an iron protein (which uses iron as a necessary catalytic cofactor), and selenium is needed because thioredoxin reductase (which is needed for electron transport to ribonucleotide reductase via thioredoxin) is a selenoprotein Citation859. The combination of dietary deficiency of sulphur amino acids and selenium, which might be geographically widespread in sub-Saharan Africa Citation551 Citation858, must therefore be expected to be strongly unfavourable, as far as DNA repair is concerned, and the situation will not be improved by simultaneous deficiency of zinc, iron or B-group vitamins also needed for normal DNA synthesis and repair.

Niacin and tryptophan are important for DNA repair not only because of the role of NADPH in both of the two parallel chains of electron transport leading via glutaredoxin and thioredoxin to ribonucleotide reductase (since thioredoxin reductase, like glutathione reductase, is a flavoprotein using NADPH as reducing cofactor), but also because of the role of NAD+ as substrate for poly(ADP-ribose) polymerase, which is an enzyme needed for normal DNA repair i.a. because it functions as an intracellular sensor for DNA strand breaks Citation860 Citation861 Citation862 . The combination of niacin and tryptophan deficiencies deficiencies leading to NAD+ depletion Citation863 is therefore mutagenic Citation864 Citation865, which should give strong reason for concern about the situation in those parts of sub-Saharan Africa where there are many poor families subsisting on monotonous diets based mainly on tryptophan-poor varieties of maize – which can easily lead to simultaneous niacin and tryptophan deficiency Citation551.

It should be noted that supplementation with niacin has been reported to have a similar protective effect as taurine supplementation against lower airway fibrosis induced by the ROS-generating anticancer drug bleomycin Citation63 Citation64 Citation66 Citation68 Citation69 Citation70 Citation71 Citation72 Citation74 Citation75 Citation76 , suggesting that high doses of niacin might have a little-noticed antioxidant protective effect. Since bleomycin has a chemical mechanism of action overlapping strongly with that of ionizing radiation because it functions in combination with iron as a generator of reactive oxygen species (ROS) which next damage the DNA molecules Citation866 Citation867 Citation868 , these observations suggest that high doses of niacin possibly also might have a similar protective effect against the fibrogenic and perhaps also other harmful effects of high doses of ionizing radiation. This is supported by the epidemiological observation that high dietary intake of niacin is associated with decreased chromosome translocation frequency in airline pilots Citation869.

Deficiencies of folate and vitamin B12 can mimic the effect of ionizing radiation in damaging DNA by causing single- and double-strand breaks, oxidative lesions, or both Citation870, but the same can also happen because of deficiencies of vitamin B6, niacin, vitamin C, vitamin E, niacin, iron or zinc Citation870. It was estimated in an article by Bruce Ames published in 1999 that the percentage of the population of the United States that had a low intake (< 50% of the RDA) for each of these eight micronutrients then ranged from 2% to more than 20% Citation870. A level of folate deficiency causing chromosome breaks occurred in approximately 10% of the population of the United States, and in a much higher percentage of the poor Citation870. His estimate was that such common micronutrient deficiencies that are likely to damage DNA by the same mechanism as for radiation and many chemicals appear to be orders of magnitude more important than the latter Citation870. However, the problem of dietary deficiencies mimicking the DNA-damaging effects of ionizing radiation must be expected to be even worse in many of the poor countries in the world than it is in the United States.

In vitro experiments indicate that genomic instability in human cells is minimized when folic acid concentration in culture medium is >227 nmol/l Citation854. Intervention studies in humans have shown: (a) that DNA hypomethylation, chromosome breaks, uracil misincorporation and micronucleus formation are minimized when red cell folate concentration is >700 nmol/l; and (b) micronucleus formation is minimized when plasma concentration of vitamin B12 is >300 pmol/l and plasma homocysteine is <7.5 micromol/l Citation854. These concentrations are achievable at intake levels in excess of current RDIs, i.e. more than 200-400 microgram folic acid per day and more than 2 microgram vitamin B12 per day Citation854. A placebo-controlled study with a dose-response suggests that based on the micronucleus index in lymphocytes, an RDI level of 700 microgram/day for folic acid and 7 microgram/day for vitamin B12 would be appropriate for genomic stability in young adults Citation854. It is thought that dietary intakes above the current RDI may be particularly important in those with extreme defects in the absorption and metabolism of these vitamins Citation854.

This must undoubtedly be highly relevant for the treatment of patients suffering from gastrointestinal damage after exposure to high doses of ionizing radiation. For minimizing the long-term mutagenic burden resulting from a given total absorbed dose of ionizing radiation, it would probably be useful to give workers who will be exposed to high doses of ionizing radiation an intramuscular injection with a large dose of methylcobalamin before exposure, or as soon as possible after they have been exposed. This can be safely done since cobalamin is very non-toxic, even to the extent that it can be safely administrated in huge doses as an antidote against cyanide poisoning.

It should be noted that fish protein concentrate type B is an excellent source of several of those nutrients that are needed for DNA synthesis and repair, but are often deficient in the diets of large groups of people both in sub-Saharan Africa and other poor countries (e.g. in the Caribbean), with the concentration of vitamin B12 being exceptionally high compared to other foods Citation550 Citation551. High-quality FPC can be produced very cheaply from cheap varieties of so-called industrial fish that comprise roughly 1/3 of the global wildfish catch, and that are now used for production of fish meal and oil that are mainly used as feed for animals (almost all of the fish meal and perhaps 70% or more of the fish oil). There are very strong reasons that this should be changed, if we want to optimize the health and minimize the population burden of unnatural mutations not only in the rich countries in the world, but also in the poor ones.

Similar multifactorial strategies for suppression of harmful inflammation with minimal suppression of useful antiviral and antibacterial immunity can hopefully be used both for treatment of severe radiation injury, hypervirulent avian influenza and SARS.

Following exposure to high doses of ionizing radiation, it will also be important to suppress the posttraumatic inflammatory overreaction that may be expected to come following severe radiation injury at the same time as immunological recovery and other tissue repair processes should be stimulated as well as possible, while an attempt should also be made to hinder fibrosis and scar tissue development as a consequence of the radiation injury.

There might be an apparent conflict between the need to suppress inflammation and the need to stimulate good immunological recovery, and anti-inflammatory preparates with too strong immunosuppressive effects, like glucocorticoid preparates, should therefore be avoided. An optimal diet may, however, be antiinflammatory i.a. because it may help to suppress prostaglandin overproduction and thus may help to reduce neurogenic inflammation Citation534, at the same time as it also facilitates immunological recovery. This is dramatically illustrated by the author's personal experience with the great value of the same type of food (viz. fish protein concentrate type B) both for treating severely ill patients suffering from the combination of malnutrition and infectious disease in Africa Citation663 and for treatment of rheumatoid arthritis in his mother, who was permanently cured for nearly 17 years until she died. It would perhaps be helpful as part of emergency preparedness efforts not only for encountering nuclear disasters, but also for reducing the number of deaths caused by pandemics with hypervirulent avian influenza or SARS, if countries all over the world (but especially those with nuclear power plants) could establish large stores with this product (FPC type B), packed under inert gas to prevent oxidative rancidification, and perhaps having it both in form of powder and granulate to make it practically easier to use it in patients.

Space does not allow any extensive discussion here of all those other substances, in addition to taurine, that might also be useful as part of multifactorial therapeutic intervention strategies following severe radiation injury either because they are radioprotective or for other reasons (e.g. because of anti-inflammatory or immune-stimulating effects). Attention may, however, be drawn to those multifactorial treatment strategies that have been proposed for hypervirulent avian influenza in earlier survey articles Citation520 Citation532, and also to a short article about “radiation biochemistry” written by the present author Citation817. For optimizing antioxidant and antinitrative protection in patients suffering from hypervirulent avian influenza, it was suggested in the articles about malignant influenza Citation520 Citation532 to use a combination of good diet and supplementation with high doses of taurine, high doses of the endogenously synthesized antioxidant coenzyme Q10 and high doses of the antioxidant and immunostimulatory hormone melatonin. A similar combination would probably be useful for patients who have been exposed to high levels of ionizing radiation as well.

The combination of optimal diet and high doses of melatonin (with strong radioprotective effects Citation711 Citation712 Citation713 Citation714 Citation715 Citation716 Citation717 Citation718 Citation719 Citation720 Citation721 Citation722 Citation723 Citation724 Citation725 Citation726 Citation727 Citation728 Citation729 Citation730 Citation731 Citation732 Citation733 Citation734 Citation735 Citation736 Citation737 Citation738 Citation739 Citation740 Citation741 Citation742 Citation743 Citation744 Citation745 Citation746 Citation747 Citation748 Citation749 Citation750 Citation751 Citation752 Citation753 Citation754 Citation755 Citation756 Citation757 Citation758 Citation759 Citation760 Citation761 Citation762 Citation763 Citation764 Citation765 Citation766 Citation767 Citation768 Citation769 Citation770 Citation771 ) would be expected to helpful not only for optimizing radioprotection, but also for improving antibacterial and antiviral immunological functions Citation520 Citation532. Melatonin exercises its immunoregulatory effects at multiple levels both in the pineal gland, in the bone marrow and directly on peripheral leukocytes, with some of the effects being direct and others indirect because melatonin stimulates the synthesis of other signal substances with immunoregulatory functions, such as thymic hormones, opioid peptide cytokines produced by cells in the bone marrow, and other cytokines Citation520 Citation532 Citation533 Citation871 Citation872 Citation873 Citation874 Citation875 Citation876 Citation877 Citation878 Citation879 Citation880 Citation881 . Melatonin does not only stimulate the production of natural killer cells, monocytes and other leukocytes, but alters the balance of T helper (Th)-1 and Th-2 cells mainly towards Th-1 responses and increases the production of relevant cytokines such as interleukin-2 (IL-2), IL-6, IL-12 and interferon-gamma Citation880.

Of special interest is the observation that high-dose melatonin supplementation protects against drug-induced leukopenia and thrombocytopenia in cancer patients who have been treated with cytotoxic drugs or interleukin-2, as well as in patients suffering from various other diseases Citation882 Citation883 Citation884 Citation885 Citation886 Citation887 Citation888 Citation889 Citation890 Citation891 . These effects are most plausibly explained by a combination of antioxidant protective effects and direct or indirect (via opoiod cytokines?) stimulating effects of melatonin on the growth of myeloid precursor cells, possibly including macrophages Citation871 Citation872 giving rise to platelets and myeloid-derived leukocytes. The latter effect might be expected to be highly relevant also for patients who suffer from leukopenia and thrombocytopenia because they have been exposed to high levels of ionizing radiation Citation892 Citation893 Citation894 Citation895 Citation896 Citation897 Citation898 Citation899 . Another substance produced by the pineal gland, viz. 5-methoxytryptamine, has also been found to have a similar effect Citation900 Citation901. Protective effects against radiation-induced leukopenia and thrombocytopenia have, moreover, also been reported for the hydroxylated dehydroepiandrosterone metabolite androstenediol Citation892 Citation893 Citation894 Citation895 and tocopherol succinate Citation897.

The combination of guanosine + inosine has also been found to be similarly protective Citation896. This observation is highly compatible with the hypothesis which has been discussed above that immunosuppression and intestinal injury following exposure to high levels of ionizing radiation may happen because of nucleotide depletion caused by a high rate of nucleotide consumption for DNA repair. If this explanation for the protective effect of the guanosine + inosine combination is correct, it would probably have been even better to give nucleoside precursors for all of the four different nucleotides that are used as building blocks during DNA synthesis and repair rather than for only two of them.

An important question concerns the safety of giving melatonin at high dosage levels to human patients (e.g. 20 to 40 mg/day for adult patients). Abundant animal experimental data suggest that melatonin given at high dosage levels might be useful in several different diseases as well as following severe trauma, but these animal experiments have in most cases not been adequately followed up by clinical studies with human patients, with the consequence that clinical experience with high-dose melatonin preparates is not very extensive, either as regards useful therapeutic effects or undesirable side effects. There are, however, some important exceptions to this, especially with the use of high-dose melatonin supplementation as adjunctive therapy for cancer, where there is a large number of studies from Lissoni and collaborators in Italy over a period of more than twenty years that involves a considerable cumulate number of patients that should be more than adequate for permitting an assessment of the frequency of adverse side effects compared to the frequency of positive ones (even though it may sometimes be difficult, when dealing with severely ill patients, to distinguish between adverse drug effects and adverse consequences of the disease itself). It would probably be useful if Lissoni himself could carry out some kind of metaanalysis of the results of his own studies, pooling the data for all those different patients who have been given high-dose melatonin over a period of more than twenty years not only concerning positive therapeutic effects, but also for putative adverse side effects, even when it is far from certain that there is any form of causal connection.

Some examples of publications from this group have been given here Citation892 Citation893 Citation894 Citation895 Citation896 Citation897 Citation898 Citation899 , but there are far more (far too numerous that they can be referenced here!) that easily can be found on PubMed by combining the search words melatonin and cancer with the author name. If there are important negative side effects of high-dose melatonin preparates, they would presumably be similar in patients suffering from other diseases as can be seen in cancer patients. But the first-glance overall impression from the clinical effects observed in a large total number of cancer patients is that the toxicity of this drug is very low (at least when compared to its positive therapeutic effects) and its safety correspondingly high, even when it is given at a dosage level of 20-40 mg/day to adult patients.

Another example of using melatonin at high dosage levels is a recent report from Spain about treatment of children suffering from Duchenne's muscular dystrophy with melatonin at at a dosage level of 70 mg/day Citation902. Ten patients aged 12.8 +/- 0.98 yr, were treated with melatonin (60 mg at 21:00 hr plus 10 mg at 09:00 hr) over 6 months, and plasma levels of lipid peroxidation (LPO), nitrites (NOx), interleukin-1beta (IL-1beta), IL-2, IL-6, tumor necrosis factor-alpha, interferon-gamma, and plasma markers of muscle injury, were determined at 3, 6 and 9 months of treatment Citation902. Healthy age- and sex-matched subjects were used as controls. The results show a significant increase in LPO, NOx, and cytokine levels in the blood plasma of Duchenne patients compared with controls Citation902. Melatonin administration reduced these values to control levels at 3 months of treatment, decreasing further 9 months later Citation902. In parallel, melatonin also reduced plasma levels of creatine kinase (50%), lactate dehydrogenase (28%), aspartate aminotransferase (28%), alanine aminotransferase (20%), and myoglobin (13%) Citation902. The authors conclude that the high-dose melatonin administration must have reduced significantly the hyperoxidative and inflammatory process in Duchenne patients, and that it must have reduced the muscle degenerative process Citation902.

The possibility of using other immunostimulating hormones in addition to melatonin (such as dehydroepiandrosterone or one of its more active hydroxylated metabolites, such as androstenediol Citation532, but also thymic hormones Citation533), should, however, also be considered not only for treatment of hypervirulent avian influenza and other dangerous viral infections, but also for promoting the recovery of immunological functions in patients who have been exposed to high doses of ionizing radiation. There is no good reason why androstenediol should be much less effective not only for reducing the lethality of infection with very dangerous viruses Citation532 Citation533, but also as a radioprotectant Citation892 Citation893 Citation894 Citation895 in human patients than has been found in animal experiments.

Similarly as earlier has been proposed for use as part of a multifactorial treatment strategy for malignant influenza (for stimulating the useful antiviral immune response without enhancing harmful inflammation) Citation532, it is possible that beta-glucans (which in this case probably would need to be orally administrated) might be valuable for stimulating recovery of the immune systems in patients who have been exposed to high doses of ionizing radiation. It is, furthermore, possible that some of those Toll-like receptor agonist preparates for stimulation of Th1 (antiviral and antitumour) immune responses that have been used for treatment of cancer patients, such as OK-432 Citation903 Citation904 also might be useful here, provided that they can be applied at a dosage level and by a route that does not lead to enhancement of harmful inflammation more than it enhances the capacity of the immune system to fight harmful bacteria and viruses.

Not only for improving antioxidant protection Citation520, but also for reducing the intensity of non-specific, but harmful inflammatory responses Citation532, it was proposed in the influenza articles to use a combination of good diet with high doses of taurine. There might be reason for hope that all these interventions (that should presumably be combined with each other for obtaining an optimal overall therapeutic response) also might be helpful for patients suffering from severe radiation injury.

Possible relevance of the protein kinase CK2-mediated growth-stimulatory effects of polyamines for repair of intestinal mucosal damage in malnourished patients and patients suffering from radiation injury.

Not mentioned in the influenza articles, but in my article about radiation biochemistry Citation817, polyamines are also potent radioprotective agents with polyamine depletion being corresponddingly harmful for cells that are exposed to ionizing radiation, with the radioprotective effect apparently being due to a combination of different mechanisms including hydroxyl radical scavenging and structural effects (both as a direct consequence of polyamine binding to the DNA molecule and conformation changes in the DNA molecule itself) that reduce the access of reactive molecules to vulnerable sites in the DNA molecule Citation905 Citation906 Citation907 Citation908 Citation909 Citation910 Citation911 Citation912 Citation913 Citation914 Citation915 Citation916 Citation917 Citation918 Citation919 . It may be relevant that polyamines, especially spermine, have been found to be highly potent antioxidants also capable of scavenging singlet oxygen Citation920 Citation921 Citation922 Citation923 Citation924 Citation925 Citation926 Citation927 Citation928 Citation929 Citation930 Citation931 Citation932 Citation933 Citation934 Citation935 Citation936 Citation937 Citation938 Citation939 ; however, they can sometimes also have prooxidant effects Citation940.

Pharmacological inhibition of polyamine synthesis in the tumour cells must be expected to lead to enhanced sensitivity to radiation therapy Citation817 Citation905 Citation906 Citation907 Citation909, but polyamine depletion will also enhance the radiation sensitivity of normal tissues adjacent to the tumour. It is, however, very common (especially in fast-growing tumour cell populations) that polyamine synthesis rates are higher in tumour cells than in normal cells, for which reason pharmacological inhibition of their synthesis might be expected to be differentially more harmful to the tumour cells than to the adjacent normal tissues, thus hopefully offering an opportunity to improve the therapeutic ratio for the radiation therapy (by enhancing damage to the tumour cells more than to adjacent normal tissues). If the uptake capacity for polyamines in the tumour cells is not too high, it should in principle be possible to use a combination of pharmacological polyamine synthesis inhibition and polyamine supplementation for reducing the polyamine concentration in the tumour cells while maintaining polyamine concentrations in normal cells at a normal level Citation817. Some other methods for hopefully improving the therapeutic ratio of radiation therapy (after the expression of superoxide dismutase and NO synthase in the tumour cells has been measured) have also been mentioned in the same article Citation817.

Polyamines are essential to cellular growth processes Citation941 Citation942 Citation943 , which may partly be explainned by their structural role in chromosomes when forming complexes with DNA. It was previously believed that polyamines are synthesized by every cell in the body when required, but it has later been shown that, as in the case of non-essential as well as essential amino acids (and in the case of conditionally essential nutrients, such as nucleotides and taurine), the diet can supply sufficient amounts of polyamines to support cell renewal and growth Citation941. The relative contribution of dietary compared to endogenously produced polyamines would be expected to be especially high in the intestinal mucosa, where the epithelial cells also have especially fast turnover and the growth rate for the progenitors of mature epithelial cells must be correspondingly high. The concentration of polyamines in human milk is also relatively high Citation944 Citation945 Citation946 Citation947 Citation948 Citation949 , about 10 times higher than in infant formula Citation946, suggesting a role of polyamines in the normal development of the intestine (and associated microflora?), and perhaps also in the normal development of the immune system in the infant.

It is therefore possible that a diet rich in polyamines or polyamine supplementation might be helpful for patients suffering from severe radiation injury, especially for stimulating enterocyte precursor growth and hence tissue repair processses in the intestinal mucosa. The importance of polyamines for the control of growth and repair processes in the intestinal mucosa as well as of the functional properties (e.g. disaccharidase expression) of the intestinal brush border is now well documented by experimental studies Citation950 Citation951 Citation952 Citation953 Citation954 Citation955 Citation956 Citation957 Citation958 Citation959 Citation960 Citation961 Citation962 Citation963 Citation964 Citation965 Citation966 Citation967 Citation968 Citation969 . It may be worth mentioning-as an example possibly illustrating the practical clinical relevance of these experimental observations-that it was a diet not only rich in taurine, but also in DNA, RNA and polyamines (from capelin that had been caught during the spawning season) that I was using myself when treating a large group of patients suffering from the combination of malnutrition, measles and diarrhoea while working for the Red Cross in Africa in 1974, with dramatically positive therapeutic results as earlier explained.

An important part of the mechanism, in addition to their role as DNA-binding structural components in chromosomes, for explaining the stimulating effect of polyamines on cellular growth processes must be their role as intracellular signal molecules (or “second messengers”) that regulate various enzymes with regulatory functions, such as protein kinase CK2 (which was earlier called casein kinase-2) Citation970 Citation971. Protein kinase CK2 regulates hundreds of proteins both in the nucleus, in the cytosol and in the plasma membrane, thus being involved in the regulation both of gene transcription, RNA processing and protein synthesis by the ribosomes. It plays a global role in activities related to cell growth, cell death, and cell survival – to an extent giving it the status of a "master regulator" in the cell Citation972. As examples of proteins associating with and/or regulated by protein kinase CK2 may be mentioned the nucleolar proteins nucleolin Citation973 Citation974, Nopp140 Citation975 and nucleolar protein B23 Citation976, topoisomerase I Citation977, insulin receptor substrate 1 (IRS-1) Citation974, the ATP-binding cassette protein ABC50 Citation978, which interacts with eIF2 (eukaryotic initiation factor 2), a protein that plays a key role in translation initiation and in its control, and in regulation of ribosomes Citation978, the von Hippel-Landau protein Citation979, which can be mutated in cancer patients, and the transcription factor NF-kappaB Citation972.

It is therefore not implausible that an important part of the explanation for the exceptionally rapid recovery of my malnourished patients with measles and diarrhoea in Tillia in 1974 could have been strong polyamine-mediated activation of protein kinase CK2 in intestinal mucosal epithelial cells and leukocytes, which may have led to enhancement of the activities of several different proteins associated with cellular growth processes and therefore to more rapid synthesis of mRNA and protein combined with more rapid cell replication, as long as there was no deficiency of any nutrient that could have led to growth limitation either among the essential ones or among those conditionally essential nutrients that mucosal epithelial cells and leukocytes need for rapid growth. It would obviously be therapeutically relevant, if it might be possible to achieve the same also in patients with severe gastrointestinal symptoms because of radiation injury.

The "master regulatory" Citation972 functions of protein kinase CK2 as a regulatory enzyme simultaneously regulating several proteins in the nucleus, in the ribosomes, in the cytoskeleton and in the plasma membrane makes it an excellent candidate to function at a very high level – as one of the “top generals” – in the hierarchy of proteins that regulate the protein synthesis-dependent anatomical changes (with strengthening of the synaptic contacts between some cell pairs and weakening of the synaptic contacts between other cell pairs) that are needed for establishment of long-term memory in the brain. It may thus be suspected that polyamines and protein kinase CK2 may be very important not only in the fast-growing intestinal mucosal cells, but also for learning processes in the brain.

Ornithine decarboxylase, which is the most important rate-limiting enzyme in the polyamine biosynthetic pathway, is expressed abundantly in the brain, but most of it is present in form of an inactive complex with the inhibitor protein antizyme Citation980, which may be regarded as an inactive precursor storage form of the active enzyme similarly as when NF-kappaB is rendered inactive by complex formation with IkappaB. The ratio of the abundance of inactive precursor to active ornithine decarboxylase in the brain might possibly be as high as about 50/1 Citation980, suggesting that the local rate of polyamine synthesis in parts of the nerve cells can be enhanced very fast following appropriate local stimulation. Since the inactive antizyme/ornithine decarboxylase complex can be dissociated by GTP, leading to activation of the enzyme Citation981, activation of nucleoside diphosphate kinases in the synaptic region might presumably be of crucial importance for initiation of this process. The nucleoside diphosphate kinases make GTP from GDP at the same time as ATP is degraded. The same mechanism for activating ornithine decarboxylase has been found also in Escherichia coli Citation982, and might presumably be phylogenetically extremely ancient.

Conclusions

It is reasonable to assume that the radioprotectant action of taurine, when given before irradiation (or a weakening of this protective effect in animals or human patients suffering from taurine depletion before the exposure to ionizing radiation), partly may be explained by its function as an important intracellular antioxidant in mammalian nucleated cells and platelets. Evidence for this antioxidant effect of taurine is mostly indirect and relies on the demonstration of a pronounced antidote effect of high doses of taurine against a large range of toxic substances that taken as a group have little in common except that the substance itself or some of its metabolites function as prooxidants in one way or another, or inhibit important antioxidative enzymes.

The mechanisms responsible for the in vivo antioxidant effects of taurine are, nevertheless, poorly understood because taurine is not particularly effective as a scavenger of free radicals or other oxidants, except hypohalite ions and aldehydes. It is therefore proposed here that it may also function by an entirely different mechanism by participating in formation of mixed complexes with iron, especially in form of complexes where the iron atom is coordinated with three oxygen atoms in phosphate groups on one side and three oxygen atoms in the sulfonic acid group of taurine on the other side, thus shielding the iron atom completely from contact with molecular oxygen, ROS, organic hydroperoxide groups or reducing organic molecules until the complex has dissociated. Experimental confirmation of this hypothesis is, nevertheless, still lacking, and the stability constants for mixed complexes of this type are also unknown. If this hypothesis is valid and applicable also for iron complex-bound with DNA molecules, it might presumably help to explain why the taurine concentration in mammalian cells (at least some of them) is especially high in the mitochondria.

Severe radiation injury must be expected to lead to a severe post-traumatic inflammatory response, similarly as happens after other forms of severe trauma (e.g. severe burns and brain stroke), and it is known from these other forms of trauma that a strong post-traumatic inflamematory response both can be associated with significantly enhanced death risk (e.g. following severe burns) and exacerbation of permanent tissue damage following the injury (which is perhaps most typically seen following brain stroke and severe mechanical trauma to the brain). Thus, it is a reasonable working hypothesis that both the death risk and extent of permanent tissue injury following severe radiation injury may be reduced by good anti-inflammatory therapy following the exposure. But since immunosuppression and damage to mucosal organs where pathogenic organisms easily can enter also are among the common consequences of severe radiation injury, the challenge to the physician is to dampen unspecific and harmful inflammatory over-response without simultaneously inhibiting too much the capacity of the immune system for good antibacterial and antiviral immune responses. Ideally, the immune system ought to be strengthened at the same time as the harmful inflammatory reactions can be inhibited as much as possible. This double challenge is very similar to the challenge also facing the doctor when trying to hinder that a patient infected with hypervirulent H5N1 avian influenza or SARS shall die from the disease, and there may be good reason for hope that a multifactorial strategy that is helpful for achieving this double objective in patients suffering from hypervirulent influenza or SARS also may be helpful for patients who have been exposed to high levels of ionizing radiation and vice versa.

Taurine may be expected to exert significant anti-inflammatory and also analgesic effects by a combination of several different mechanisms: 1) reducing prostaglandin synthesis and PKC oxidative activation because of its antioxidant effects and therefore helping to reduce C-fiber-mediated neurogenic inflammation, 2) functioning as an agonist ligand of inhibitory glycine receptors on neutrophil granulocytes and macrophages (and perhaps on eosinophils and mast cells as well?), 3) functioning as an agonist ligand of inhibitory receptors on C-fibres and thus reducing neurogenic inflammation, 4) reacting with hypohalite ions to form taurine chloramine and taurine bromamine, which inhibit activation of the transcripton factor NF-kappaB and therefore may help to reduce the expression of a large number of proinflammatory proteins, 5) regulating the rate of membrane transport of calcium in ways that would be expected to contribute to reduction of the secretion of histamine and other proinflammatory substances from storage granula in leukocytes and also reduce the rate of leukotriene synthesis.

The anti-inflammatory and analgesic effects of taurine may be expected to be therapeutically useful in several different diseases both when harmful inflammation develops as a consequence of infection (as in hypervirulent influenza or SARS, but most likely also in various diarrhoeal diseases) and when it has a non-infectious cause, as in rheumatoid arthritis, in asthma and other allergic diseases and after ischemia (e.g. myocardial infarction), and also following severe trauma (both severe burn injuries and severe mechanical truma) for reducing the risk that the patient shall die from shock or multiorgan failure. They may also be expected to be relevant in various common pain conditions, especially when the pain develops a consequence of ischemia, e.g. because of abnormal static loads or spasms in skeletal muscle (and perhaps also during childbirth), in which case the antiischemic protective effect of taurine also may be relevant.

In allergic diseases, the rate of production of taurine bromamine must be expected to depend on the concentration of bromide in blood plasma, which is most likely often strongly reduced in humans compared to the natural level found in wild animals because of high consumption in the human population of table salt with a Br/Cl ratio much lower than in seawater. If the plasma bromide concentrations of asthma patients could be normalized to the same level that is found in wild animals, taurine therapy would be expected to have considerably better effect than if bromide deficiency is not corrected.

For rheumatoid arthritis, a case history is discussed for illustrating the possible clinical relevance of the anti-inflammatory effects of taurine. A hypothesis for explaining major features in the pathogenesis of rheumatoid arthritis that represents a more elaborate version of one first proposed by Hajizadeh et al. Citation552 is presented as background for discussion of possible mechanisms explaining why the patient was permanently cured after she had started to take high daily doses of fish powder as a source of selenium – but the product is also a good source of taurine, glycine and other nutrients with anti-inflammatory effects. It is proposed that a vicious circle develops where a combination of hypoxia and much NO and proinflammatory cytokine production leads to necrotic cell death, while proinflammatory substances released when cells die in necrosis in combination with neurogenic inflammation cause enhancement of the production of NO and proinflammatory cytokines, such as TNF-alpha, which leads to enhancement of the number of cells dying through necrosis and therefore more inflammation. Taurine may, however, help to break this vicious circle both because of the direct inhibitory effects of taurine itself and taurine chloramine on neutrophils and macrophages and because of C-fibre inhibition.

Taurine is reported to function sometimes as a factor enhancing the rate of cell growth processes, especially in leukocytes and nerve cells, and sometimes as a growth-inhibitory factor. These opposite effects are probably due to very different mechanisms, with the growth inhibitory effects of taurine being a consequence of its interference with important intracellular signal systems regulating cell growth, while the growth-stimulating effects of taurine might be a consequence of its role as a nutrient because of its function as one of the major intracellular osmolytes and because of the mechanisms connecting cell volume regulation directly with the regulation of the rates of protein synthesis and protein degradation-with enhancement of the volume of the cell leading to enhanced protein synthesis and reduced protein degradation, while shrinkage of the cell has opposite effects. Considering the opposite regulatory effects of taurine on the growth of different cell types, there is good reason to hope that taurine supplementation in a patient that has become taurine-depleted because of radiation injury will help the immune system to faster recovery at the same time as it may help to hinder the development of fibrosis (which would be similar to the well-documented anti-fibrotic effect of taurine in the lower airways of animals exposed to the anticancer drug bleomycin).

Taurine should not be regarded as a “magic bullet” that can be used alone for prevention or treatment of severe radiation injury. Rather, it should be used as one among several components in multifactorial prophylactic and therapeutic interventions with multiple aims, viz. Citation1 optimizing cellular antioxidant defense, Citation2 optimizing DNA repair during and following the exposure to high doses of ionizing radiation, Citation3 minimizing the post-traumatic inflammatory reaction, Citation4 optimizing the recovery of immunological functions, Citation5 optimizing all other forms of tissue repair and regeneration, especially in the gastrointestinal tract (if at all feasible), and Citation6 preventing the development of fibrosis and scar formation.

Something similar may also be expected to be the case in several other disease conditions where taurine supplementation might be therapeutically useful, but where the best therapeutic effect probably can be obtained not by using taurine alone (not even when given at a very high daily dosage level), but by using it as part of a multifactorial treatment strategy, e.g. in combination with other nutrients also having antiinflammatory, antioxidative or antiischemic protective effects.

A practical problem associated with the use of taurine as an anti-inflammatory or analgesic agent is its rapid urinary excretion when applied in large doses. It would probably be better to use slow-release formulations rather than such rapidly absorbed taurine preparates that are now available in the market. Since it is well-known by the pharmaceutical industry how slow-release drug formulations can be made, it should not be practically infeasible (or much expensive) to produce in a very short period of time (if ordered by Japanese health authorities) as large quantities as might be needed for such preparates in the current emergency situation.

For improving emergency preparedness against future nuclear accidents in Japan or other countries, it will nevertheless be important to test the clinical effect of such preparates as well as possible under more normal circumstances, i.e. in non-emergency situations.

Something similar can be said also about melatonin, which has a fast turnover not because of renal excretion, but because of enzymatic degradation related to its function as a hormone helping to control diurnal biological rhythms (for which reason the blood concentration shall rapidly go down in the morning when light comes and people wake up). High-dose slow-release formulations should probably be preferred also here, but they need to be tested in non-emergency situations for verification that they indeed have the sought-for protective effects.

If such slow-release, high-dose taurine and melatonin preparates are found to have the desired effect, stockpiles should be made all over the world not only for improving emergency preparedness for the next large nuclear power plant catastrophe, but also for pandemics with highly lethal viruses, e.g. hypervirulent avian influenza or a new form of SARS virus that is not only more infective than the previous one, but also more lethal (similarly as the wave 2 Spanish Flu virus was both more infective and more lethal than the wave 1 Spanish Flu virus).

For pandemic emergency preparedness, it is also important to have enough food stockpiled in order to make it practically feasible to impose drastic measures including house quarantine to minimize geographic dispersal of the dangerous virus Citation532 Citation983. Such food stores for improving disaster preparedness (which might well include fish protein concentrate type B) might, however, also be useful after severe nuclear disasters by securing an adequate supply of foods that have not been radioactively contaminated, and could thus serve a double purpose (at the same time as they would, of course, also be helpful for minimizing the number of deaths following severe weather catastrophes, as in the hypothetical case that Yellowstone should blow up with one of its 1 per 700 000 years frequency mega-eruptions).

It should probably be useful to stockpile fish protein concentrate B (FPC type B) as part of such emergency preparedness stores because of its special value both for immunonutrition and as part of multiple anti-inflammatory therapy, at the same time as it is a highly concentrated product that can be produced and transported cheaply. It should then be stored in rock shelters under nitrogen to prevent oxidative rancidification, under suitable humidity conditions and at low temperature to make it possible to store the product over very long periods of time without any form of quality degradation.

The chemical mechanisms causing cellular damage during exposure to ionizing radiation and during ischemia followed by reperfusion are in large measure the same. It is therefore not surprising that several substances have been found in animal experiments to be protective in both situations. But much of the knowledge that is now available from this from a very large number of studies with several different substances in many different organs and species is not practically utilised in clinical medicine. The health economic benefits for society if this could be changed are potentially enormous, notably for acute therapy of cerebral stroke and myocardial infarction.

References

  • Abe M, Takahashi M, Takeuchi K, Fukuda M. Studies on the significance of taurine in radiation injury. Radiation Res. 1968; 33: 563–73.
  • Sugahara T, Nagata H, Tanaka T. [Experimental studies on radiation protection by taurine]. [Article in Japanese]. Nippon Igaku Hoshasen Gakkai Zasshi. 1969; 29: 156–61.
  • Kolesnikov IuA, Novosel'tseva SD, Iartsev EI, Kudriashov IuB, Bogatyrev GP. [Antiradiation properties of taurine compounds]. [Article in Russian]. Radiobiologiia. 1975;15: 928–31.
  • Feuer L, Benkó G. Effect of glutaurine and its derivatives and their combinations with radiation protective substances upon irradiated mice. Acta Radiol Oncol. 1981; 20: 319–24.
  • Feuer L, Ormai S. Effect of protein-free parathyroid extract (PF-PTE) and gamma-L-glutamyl-taurine (glutaurine) on X-ray induced hyperglycaemia in the rat. Acta Med Acad Sci Hung. 1981; 38: 159–62.
  • Mashkova NIu, Alekseeva EN, Dokshina GA. [Possible regulation by taurine of the intracellular potassium levels in hepatocytes and erythrocytes of irradiated rats]. [Article in Russian]. Radiobiologiia. 1983; 23: 758–60.
  • Mashkova NIu, Borovikova GV, Dokshina GA. [Effect of insulin and taurine on the potassium content of the perfused liver in irradiated rats]. [Article in Russian]. Radiobiologiia. 1983; 23: 240–3.
  • Mashkova NIu, Alekseeva EN, Dokshina GA. [Effect of taurine on the potassium content of rat thymocytes after irradiation]. [Article in Russian]. Radiobiologiia. 1987; 27: 372–4.
  • Robb WB, Condron C, Moriarty M, Walsh TN, Bouchier-Hayes DJ. Taurine attenuates radiation-induced lung fibrosis in C57/Bl6 fibrosis prone mice. Ir J Med Sci. 2010; 179: 99–105.
  • Bigwood EJ. [On the increased excretion of taurine and beta aminoisobutyric acid in urine following irradiation and the sensitivity of these biochemical reactions]. [Article in German]. Strahlenschutz Forsch Prax. 1964; 4: 183–97.
  • Fromageot P, Boquet PL. [On the origin of the increased taurine excretion in irradiated rats]. [Article in German]. Strahlenschutz Forsch Prax. 1964; 4: 199–230.
  • Streffer C, Melching HJ, Mattausch H. [Studies on biologic radiation protection. 70. On the excretion of taurine in white mice following whole body irradiation and exposure to radiation-protective substances]. [Article in German]. Strahlentherapie. 1966; 130: 146–56.
  • Pentz EI. Adaptation of the Rimini-Schryver reaction for the measurement of allantoin in urine to the autoanalyzer: allantoin and taurine excretion following neutron irradiation. Anal Biochem. 1969; 27: 333–42.
  • Streffer C, Akinsanya O, Schafferus S. [Research on the increase in taurine excretion after irradiation in mice]. [Article in German]. Strahlentherapie. 1969; 138: 733–8.
  • Raghavan KG, Nadkarni GB. Formation and excretion of taurine in x-irradiated rats. Int J Radiat Biol Relat Stud Phys Chem Med. 1970; 18: 41–9.
  • Dilley JV. The origin of urinary taurine excretion during chronic radiation injury. Radiat Res. 1972; 50: 191–6.
  • Bezkrovnaia LA, Lapteva TA, Dokshina GA, Baranova MI. [Mechanisms of taurine hyperexcretion following whole-body irradiation]. [Article in Russian]. Radiobiologiia. 1976; 16: 683–6.
  • Bezkrovnaia LA, Dokshina GA. [Sources of taurine hyperexcretion in rats after irradiation]. [Article in Russian]. Radiobiologiia. 1980; 20: 455–9.
  • Moroz BB, Vasil'ev PS, Fedorovskiı˘ LL, Grozdov SP, Morozova NV. [Effect of local x-ray irradiation of the abdominal area on the amino acid content of the blood plasma and their urinary excretion in dogs and rats]. [Article in Russian]. Radiobiologiia. 1987; 27: 332–8.
  • Bezkrovnaia LA, Kostesha NIa. [The diagnostic value of the status of urinary taurine in the early stages following the irradiation of animals]. [Article in Russian]. Med Radiol (Mosk). 1990; 35: 23–5.
  • Tyburski JB, Patterson AD, Krausz KW, Slavík J, Fornace AJ Jr, Gonzalez FJ, Idle JR. Radiation metabolomics. 1. Identification of minimally invasive urine biomarkers for gamma-radiation exposure in mice. Radiat Res. 2008; 170: 1–14.
  • Desai TK, Maliakkal J, Kinzie JL, Ehrinpreis MN, Luk GD, Cejka J. Taurine deficiency after intensive chemotherapy and/or radiation. Am J Clin Nutr. 1992; 55: 708–11.
  • Kerai MD, Waterfield CJ, Kenyon SH, Asker DS, Timbrell JA. Taurine: protective properties against ethanol-induced hepatic steatosis and lipid peroxidation during chronic ethanol consumption in rats. Amino Acids. 1998; 15: 53–76.
  • Kerai MD, Waterfield CJ, Kenyon SH, Asker DS, Timbrell JA. Reversal of ethanol-induced hepatic steatosis and lipid peroxidation by taurine: a study in rats. Alcohol Alcohol. 1999; 34: 529–41.
  • Xie Y, Li XP, Wang CW, Huang DQ, Zhu JQ, Zhang KH, Chen J. [Ethanol-induced gastric mucosal injury and the protection of taurine against the injury in rats]. [Article in Chinese]. Sheng Li Xue Bao. 1999; 51: 310–4.
  • Bleich S, Degner D. Reversal of ethanol-induced hepatic steatosis and lipid peroxidation by taurine: a study in rats. Alcohol Alcohol. 2000; 35: 215.
  • Harada H, Kitazaki K, Tsujino T, Watari Y, Iwata S, Nonaka H, Hayashi T, Takeshita T, Morimoto K, Yokoyama M. Oral taurine supplementation prevents the development of ethanol-induced hypertension in rats. Hypertens Res. 2000; 23: 277–84.
  • Kerai MD, Waterfield CJ, Kenyon SH, Asker DS, Timbrell JA. The effect of taurine depletion by beta-alanine treatment on the susceptibility to ethanol-induced hepatic dysfunction in rats. Alcohol Alcohol. 2001; 36: 29–38.
  • Balkan J, Kanbağli O, Aykaç-Toker G, Uysal M. Taurine treatment reduces hepatic lipids and oxidative stress in chronically ethanol-treated rats. Biol Pharm Bull. 2002; 25: 1231–3.
  • Erman F, Balkan J, Cevikbaş U, Koçak-Toker N, Uysal M. Betaine or taurine administration prevents fibrosis and lipid peroxidation induced by rat liver by ethanol plus carbon tetrachloride intoxication. Amino Acids. 2004; 27: 199–205.
  • Pushpakiran G, Mahalakshmi K, Anuradha CV. Taurine restores ethanol-induced depletion of antioxidants and attenuates oxidative stress in rat tissues. Amino Acids. 2004; 27: 91–6.
  • Pushpakiran G, Mahalakshmi K, Anuradha CV. Protective effects of taurine on glutathione and glutathione-dependent enzymes in ethanol-fed rats. Pharmazie. 2004; 59: 869–72.
  • Pushpakiran G, Mahalakshmi K, Viswanathan P, Anuradha CV. Taurine prevents ethanol-induced alterations in lipids and ATPases in rat tissues. Pharmacol Rep. 2005; 57: 578–87.
  • Choi MJ, Kim MJ, Chang KJ. The effect of dietary taurine supplementation on plasma and liver lipid concentrations and mineral metabolism in rats fed alcohol. Adv Exp Med Biol. 2006; 583: 243–50.
  • Park T, Cho K, Park SH, Lee DH, Kim HW. Taurine normalizes blood levels and urinary loss of selenium, chromium, and manganese in rats chronically consuming alcohol. Adv Exp Med Biol. 2009; 643: 407–14.
  • Wu G, Yang J, Sun C, Luan X, Shi J, Hu J. Effect of taurine on alcoholic liver disease in rats. Amino Acids. 2009; 36: 457–64.
  • Wu G, Yang J, Sun C, Luan X, Shi J, Hu J. Effect of taurine on alcoholic liver disease in rats. Adv Exp Med Biol. 2009; 643: 313–22.
  • Yang HT, Chien YW, Tsen JH, Chang CC, Chang JH, Huang SY. Taurine supplementation improves the utilization of sulfur-containing amino acids in rats continually administrated alcohol. J Nutr Biochem. 2009; 20: 132–9.
  • Lakshmi Devi S, Anuradha CV. Mitochondrial damage, cytotoxicity and apoptosis in iron-potentiated alcoholic liver fibrosis: amelioration by taurine. Amino Acids. 2010; 38: 869–79.
  • Bruns H, Watanpour I, Gebhard MM, Flechtenmacher C, Galli U, Schulze-Bergkamen H, Zorn M, Büchler MW, Schemmer P. Glycine and taurine equally prevent fatty livers from Kupffer cell dependent injury: an in vivo microscopy study. Microcirculation. 2010 Dec 22. 10.3402/mehd.v23i0.14787. [Epub ahead of print].
  • Waters E, Wang JH, Redmond HP, Wu QD, Kay E, Bouchier-Hayes D. Role of taurine in preventing acetaminophen-induced hepatic injury in the rat. Am J Physiol Gastrointest Liver Physiol. 2001; 280: G1274–9.
  • Acharya M, Lau-Cam CA. Comparison of the protective actions of N-acetylcysteine, hypotaurine and taurine against acetaminophen-induced hepatotoxicity in the rat. J Biomed Sci. 2010;17(Suppl 1): S35.
  • Das J, Ghosh J, Manna P, Sil PC. Acetaminophen induced acute liver failure via oxidative stress and JNK activation: protective role of taurine by the suppression of cytochrome P450 2E1. Free Radic Res. 2010; 44: 340–55.
  • Das J, Ghosh J, Manna P, Sil PC. Taurine protects acetaminophen-induced oxidative damage in mice kidney through APAP urinary excretion and CYP2E1 inactivation. Toxicology. 2010; 269: 24–34.
  • Erdem A, Gündoğan NU, Usubütün A, Kilinç K, Erdem SR, Kara A, Bozkurt A. The protective effect of taurine against gentamicin-induced acute tubular necrosis in rats. Nephrol Dial Transplant. 2000; 15: 1175–82.
  • Eldin AA, Shaheen AA, Abd Elgawad HM, Shehata NI. Protective effect of taurine and quercetin against renal dysfunction associated with the combined use of gentamycin and diclofenac. Indian J Biochem Biophys. 2008; 45: 332–40.
  • Liu HY, Chi FL, Gao WY. Taurine attenuates aminoglycoside ototoxicity by inhibiting inducible nitric oxide synthase expression in the cochlea. Neuroreport. 2008; 19: 117–20.
  • Hagar HH. The protective effect of taurine against cyclosporine A-induced oxidative stress and hepatotoxicity in rats. Toxicol Lett. 2004; 151: 335–43.
  • Hagar HH, El Etter E, Arafa M. Taurine attenuates hypertension and renal dysfunction induced by cyclosporine A in rats. Clin Exp Pharmacol Physiol. 2006; 33: 189–96.
  • Saad SY, Al-Rikabi AC. Protection effects of taurine supplementation against cisplatin-induced nephrotoxicity in rats. Chemotherapy. 2002; 48: 42–8.
  • Sato S, Yamate J, Saito T, Hosokawa T, Saito S, Kurasaki M. Protective effect of taurine against renal interstitial fibrosis of rats induced by cisplatin. Naunyn Schmiedebergs Arch Pharmacol. 2002; 365: 277–83.
  • Liao Y, Lu X, Lu C, Li G, Jin Y, Tang H. Selection of agents for prevention of cisplatin-induced hepatotoxicity. Pharmacol Res. 2008; 57: 125–31.
  • Han X, Chesney RW. Mechanism of TauT in protecting against cisplatin-induced kidney injury (AKI). Adv Exp Med Biol. 2009; 643: 105–12.
  • Han X, Yue J, Chesney RW. Functional TauT protects against acute kidney injury. J Am Soc Nephrol. 2009; 20: 1323–32.
  • Azuma J, Hamaguchi T, Ohta H, Takihara K, Awata N, Sawamura A, Harada H, Tanaka Y, Kishimoto S. Calcium overload-induced myocardial damage caused by isoproterenol and by adriamycin: possible role of taurine in its prevention. Adv Exp Med Biol. 1987; 217: 167–79.
  • Hamaguchi T, Azuma J, Awata N, Ohta H, Takihara K, Harada H, Kishimoto S, Sperelakis N. Reduction of doxorubicin-induced cardiotoxicity in mice by taurine. Res Commun Chem Pathol Pharmacol. 1988; 59: 21–30.
  • Hamaguchi T, Azuma J, Harada H, Takahashi K, Kishimoto S, Schaffer SW. Protective effect of taurine against doxorubicin-induced cardiotoxicity in perfused chick hearts. Pharmacol Res. 1989; 21: 729–34.
  • Harada H, Cusack BJ, Olson RD, Stroo W, Azuma J, Hamaguchi T, Schaffer SW. Taurine deficiency and doxorubicin: interaction with the cardiac sarcolemmal calcium pump. Biochem Pharmacol. 1990; 39: 745–51.
  • Venkatesan N, Venkatesan P, Karthikeyan J, Arumugam V. Protection by taurine against adriamycin-induced proteinuria and hyperlipidemia in rats. Proc Soc Exp Biol Med. 1997; 215: 158–64.
  • Huang XM, Zhu WH, Kang ML. Study on the effect of doxorubicin on expressions of genes encoding myocardial sarcoplasmic reticulum Ca2 +  transport proteins and the effect of taurine on myocardial protection in rabbits. J Zhejiang Univ Sci. 2003; 4: 114–20.
  • Ito T, Muraoka S, Takahashi K, Fujio Y, Schaffer SW, Azuma J. Beneficial effect of taurine treatment against doxorubicin-induced cardiotoxicity in mice. Adv Exp Med Biol. 2009; 643: 65–74.
  • Das J, Ghosh J, Manna P, Sil PC. Taurine suppresses doxorubicin-triggered oxidative stress and cardiac apoptosis in rat via up-regulation of PI3-K/Akt and inhibition of p53, p38-JNK. Biochem Pharmacol. 2011; 81: 891–909.
  • Wang QJ, Giri SN, Hyde DM, Li C. Amelioration of bleomycin-induced pulmonary fibrosis in hamsters by combined treatment with taurine and niacin. Biochem Pharmacol. 1991; 42: 1115–22.
  • Giri SN, Wang Q. Taurine and niacin offer a novel therapeutic modality in prevention of chemically-induced pulmonary fibrosis in hamsters. Adv Exp Med Biol. 1992; 315: 329–40.
  • Gordon RE, Heller RF, Heller RF. Taurine protection of lungs in hamster models of oxidant injury: a morphologic time study of paraquat and bleomycin treatment. Adv Exp Med Biol. 1992; 315: 319–28.
  • Wang Q, Hyde DM, Giri SN. Abatement of bleomycin-induced increases in vascular permeability, inflammatory cell infiltration, and fibrotic lesions in hamster lungs by combined treatment with taurine and niacin. Lab Invest. 1992; 67: 234–42.
  • Bhat M, Rojanasakul Y, Weber SL, Ma JY, Castranova V, Banks DE, Ma JK. Fluoromicroscopic studies of bleomycin-induced intracellular oxidation in alveolar macrophages and its inhibition by taurine. Environ Health Perspect. 1994; 102(Suppl 10): 91–6.
  • Blaisdell RJ, Schiedt MJ, Giri SN. Dietary supplementation with taurine and niacin prevents the increase in lung collagen cross-links in the multidose bleomycin hamster model of pulmonary fibrosis. J Biochem Toxicol. 1994; 9: 79–86.
  • Giri SN, Blaisdell R, Rucker RB, Wang Q, Hyde DM. Amelioration of bleomycin-induced lung fibrosis in hamsters by dietary supplementation with taurine and niacin: biochemical mechanisms. Environ Health Perspect. 1994; 102(Suppl 10): 137–47.
  • Blaisdell RJ, Giri SN. Mechanism of antifibrotic effect of taurine and niacin in the multidose bleomycin-hamster model of lung fibrosis: inhibition of lysyl oxidase and collagenase. J Biochem Toxicol. 1995; 10: 203–10.
  • Gurujeyalakshmi G, Iyer SN, Hollinger MA, Giri SN. Procollagen gene expression is down-regulated by taurine and niacin at the transcriptional level in the bleomycin hamster model of lung fibrosis. J Pharmacol Exp Ther. 1996; 277: 1152–7.
  • Gurujeyalakshmi G, Hollinger MA, Giri SN. Regulation of transforming growth factor-beta1 mRNA expression by taurine and niacin in the bleomycin hamster model of lung fibrosis. Am J Respir Cell Mol Biol. 1998; 18: 334–42.
  • Giri SN, Gurujeyalakshmi G, Wang Y. Suppression of bleomycin-induced increased production of nitric oxide and NF-kB activation by treatment with taurine and niacin. Adv Exp Med Biol. 2000; 483: 545–61.
  • Gurujeyalakshmi G, Wang Y, Giri SN. Suppression of bleomycin-induced nitric oxide production in mice by taurine and niacin. Nitric Oxide. 2000; 4: 399–411.
  • Gurujeyalakshmi G, Wang Y, Giri SN. Taurine and niacin block lung injury and fibrosis by down-regulating bleomycin-induced activation of transcription nuclear factor-kappaB in mice. J Pharmacol Exp Ther. 2000; 293: 82–90.
  • Giri SN. The combined treatment with taurine and niacin blocks the bleomycin-induced activation of nuclear factor-kappaB and lung fibrosis. Adv Exp Med Biol. 2003; 526: 381–94.
  • Schuller-Levis GB, Gordon RE, Wang C, Park E. Taurine reduces lung inflammation and fibrosis caused by bleomycin. Adv Exp Med Biol. 2003; 526: 395–402.
  • Schuller-Levis G, Gordon RE, Wang C, Park SY, Park E. Protection of bleomycin-induced fibrosis and inflammation by taurine. Int Immunopharmacol. 2009; 9: 971–7.
  • Cetiner M, Sener G, Sehirli AO, Ekşioğlu-Demiralp E, Ercan F, Sirvanci S, Gedik N, Akpulat S, Tecimer T, Yeğen BC. Taurine protects against methotrexate-induced toxicity and inhibits leukocyte death. Toxicol Appl Pharmacol. 2005; 209: 39–50.
  • Pierson HF, Fisher JM, Rabinovitz M. Modulation by taurine of the toxicity of taumustine, a compound with antitumor activity. J Natl Cancer Inst. 1985; 75: 905–9.
  • Finnegan NM, Redmond HP, Bouchier-Hayes DJ. Taurine attenuates recombinant interleukin-2-activated, lymphocyte-mediated endothelial cell injury. Cancer. 1998; 82: 186–99.
  • Abdih H, Kelly CJ, Bouchier-Hayes D, Barry M, Kearns S. Taurine prevents interleukin-2-induced acute lung injury in rats. Eur Surg Res. 2000; 32: 347–52.
  • Finnegan N, Toomey D, Condron C, Redmond HP, Da Costa M, Bouchier-Hayes DJ. Potentiation of the therapeutic index of interleukin-2 immunotherapy by combination with taurine in a syngeneic murine tumour model. Ir J Med Sci. 2002; 171: 85–8.
  • Maher SG, Condron CE, Bouchier-Hayes DJ, Toomey DM. Taurine attenuates CD3/interleukin-2-induced T cell apoptosis in an in vitro model of activation-induced cell death (AICD). Clin Exp Immunol. 2005; 139: 279–86.
  • Tabassum H, Rehman H, Banerjee BD, Raisuddin S, Parvez S. Attenuation of tamoxifen-induced hepatotoxicity by taurine in mice. Clin Chim Acta. 2006; 370: 129–36.
  • Tabassum H, Parvez S, Rehman H, Dev Banerjee B, Siemen D, Raisuddin S. Nephrotoxicity and its prevention by taurine in tamoxifen induced oxidative stress in mice. Hum Exp Toxicol. 2007; 26: 509–18.
  • Parvez S, Tabassum H, Banerjee BD, Raisuddin S. Taurine prevents tamoxifen-induced mitochondrial oxidative damage in mice. Basic Clin Pharmacol Toxicol. 2008; 102: 382–7.
  • Wang Q, Hollinger MA, Giri SN. Attenuation of amiodarone-induced lung fibrosis and phospholipidosis in hamsters by taurine and/or niacin treatment. J Pharmacol Exp Ther. 1992; 262: 127–32.
  • Ohta H, Azuma J, Onishi S, Awata N, Takihara K, Kishimoto S. Protective effect of taurine against isoprenaline-induced myocardial damage. Basic Res Cardiol. 1986; 81: 473–81.
  • Ohta H, Azuma J, Awata N, Hamaguchi T, Tanaka Y, Sawamura A, Kishimoto S, Sperelakis N. Mechanism of the protective action of taurine against isoprenaline induced myocardial damage. Cardiovasc Res. 1988; 22: 407–13.
  • Shi YR, Bu DF, Qi YF, Gao L, Jiang HF, Pang YZ, Tang CS, Du JB. Dysfunction of myocardial taurine transport and effect of taurine supplement in rats with isoproterenol-induced myocardial injury. Acta Pharmacol Sin. 2002; 23: 910–8.
  • Shiny KS, Kumar SH, Farvin KH, Anandan R, Devadasan K. Protective effect of taurine on myocardial antioxidant status in isoprenaline-induced myocardial infarction in rats. J Pharm Pharmacol. 2005; 57: 1313–7.
  • Yamauchi-Takihara K, Azuma J, Kishimoto S. Taurine protection against experimental arterial calcinosis in mice. Biochem Biophys Res Commun. 1986; 140: 679–83.
  • Sener G, Ozer Sehirli A, Ipçi Y, Cetinel S, Cikler E, Gedik N, Alican I. Taurine treatment protects against chronic nicotine-induced oxidative changes. Fundam Clin Pharmacol. 2005; 19: 155–64.
  • Sener G, Sehirli O, Ipçi Y, Cetinel S, Cikler E, Gedik N, Alican I. Protective effects of taurine against nicotine-induced oxidative damage of rat urinary bladder and kidney. Pharmacology. 2005; 74: 37–44.
  • Ulrich-Merzenich G, Zeitler H, Vetter H, Bhonde RR. Protective effects of taurine on endothelial cells impaired by high glucose and oxidized low density lipoproteins. Eur J Nutr. 2007; 46: 431–8.
  • Hwang DF, Hour JL, Cheng HM. Effect of taurine on toxicity of oxidized fish oil in rats. Food Chem Toxicol. 2000; 38: 585–91.
  • Tokunaga H, Yoneda Y, Kuriyama K. Protective actions of taurine against streptozotocin-induced hyperglycemia. Biochem Pharmacol. 1979; 28: 2807–11.
  • Trachtman H, Futterweit S, Bienkowski RS. Taurine prevents glucose-induced lipid peroxidation and increased collagen production in cultured rat mesangial cells. Biochem Biophys Res Commun. 1993; 191: 759–65.
  • Trachtman H, Futterweit S, Prenner J, Hanon S. Antioxidants reverse the antiproliferative effect of high glucose and advanced glycosylation end products in cultured rat mesangial cells. Biochem Biophys Res Commun. 1994; 199: 346–52.
  • Ha H, Yu MR, Kim KH. Melatonin and taurine reduce early glomerulopathy in diabetic rats. Free Radic Biol Med. 1999; 26: 944–50.
  • Wu QD, Wang JH, Fennessy F, Redmond HP, Bouchier-Hayes D. Taurine prevents high-glucose-induced human vascular endothelial cell apoptosis. Am J Physiol. 1999; 277: C1229–38.
  • Verzola D, Bertolotto MB, Villaggio B, Ottonello L, Dallegri F, Frumento G, Berruti V, Gandolfo MT, Garibotto G, Deferran G. Taurine prevents apoptosis induced by high ambient glucose in human tubule renal cells. J Investig Med. 2002; 50: 443–51.
  • Haber CA, Lam TK, Yu Z, Gupta N, Goh T, Bogdanovic E, Giacca A, Fantus IG. N-acetylcysteine and taurine prevent hyperglycemia-induced insulin resistance in vivo: possible role of oxidative stress. Am J Physiol Endocrinol Metab. 2003; 285: E744–53.
  • Han J, Bae JH, Kim SY, Lee HY, Jang BC, Lee IK, Cho CH, Lim JG, Suh SI, Kwon TK, Park JW, Ryu SY, Ho WK, Earm YE, Song DK. Taurine increases glucose sensitivity of UCP2-overexpressing beta-cells by ameliorating mitochondrial metabolism. Am J Physiol Endocrinol Metab. 2004; 287: E1008–18.
  • Mao DW, Zhao YP, Li SR, Che JH, Tan WH. [Study of teratogenicity of hyperglycemia on neural tube defects and antagonistic effect of taurine]. [Article in Chinese]. Zhonghua Fu Chan Ke Za Zhi. 2004; 39: 169–72.
  • Casey RG, Gang C, Joyce M, Bouchier-Hayes DJ. Taurine attenuates acute hyperglycaemia-induced endothelial cell apoptosis, leucocyte-endothelial cell interactions and cardiac dysfunction. J Vasc Res. 2007; 44: 31–9.
  • Derlacz RA, Sliwinska M, Piekutowska A, Winiarska K, Drozak J, Bryla J. Melatonin is more effective than taurine and 5-hydroxytryptophan against hyperglycemia-induced kidney-cortex tubules injury. J Pineal Res. 2007; 42: 203–9.
  • Huang JS, Chuang LY, Guh JY, Huang YJ, Hsu MS. Antioxidants attenuate high glucose-induced hypertrophic growth in renal tubular epithelial cells. Am J Physiol Renal Physiol. 2007; 293: F1072–82.
  • Son HY, Kim H, H Kwon Y. Taurine prevents oxidative damage of high glucose-induced cataractogenesis in isolated rat lenses. J Nutr Sci Vitaminol (Tokyo). 2007; 53: 324–30.
  • Anuradha CV, Balakrishnan SD. Taurine attenuates hypertension and improves insulin sensitivity in the fructose-fed rat, an animal model of insulin resistance. Can J Physiol Pharmacol. 1999; 77: 749–54.
  • Anitha Nandhini AT, Balakrishnan SD, Anuradha CV. Taurine modulates antioxidant potential and controls lipid peroxidation in the aorta of high fructose-fed rats. J Biochem Mol Biol Biophys. 2002; 6: 129–33.
  • Nandhini AT, Balakrishnan SD, Anuradha CV. Response of liver antioxidant system to taurine in rats fed high fructose diet. Indian J Exp Biol. 2002; 40: 1016–9.
  • Harada H, Tsujino T, Watari Y, Nonaka H, Emoto N, Yokoyama M. Oral taurine supplementation prevents fructose-induced hypertension in rats. Heart Vessels. 2004; 19: 132–6.
  • Nandhini AT, Anuradha CV. Hoe 140 abolishes the blood pressure lowering effect of taurine in high fructose-fed rats. Amino Acids. 2004; 26: 299–303.
  • Nandhini AT, Thirunavukkarasu V, Anuradha CV. Potential role of kinins in the effects of taurine in high-fructose-fed rats. Can J Physiol Pharmacol. 2004; 82: 1–8.
  • Nandhini AT, Thirunavukkarasu V, Anuradha CV. Taurine modifies insulin signaling enzymes in the fructose-fed insulin resistant rats. Diabetes Metab. 2005; 31: 337–44.
  • Nandhini AT, Thirunavukkarasu V, Anuradha CV. Taurine prevents collagen abnormallities in high fructose-fed rats. Indian J Med Res. 2005; 122: 171–7.
  • Nandhini TA, Thirunavukkarasu V, Ravichandran MK, Anuradha CV. Taurine prevents fructose-diet induced collagen abnormalities in rat skin. J Diabetes Complications. 2005; 19: 305–11.
  • Nandhini AT, Thirunavukkarasu V, Ravichandran MK, Anuradha CV. Effect of taurine on biomarkers of oxidative stress in tissues of fructose-fed insulin-resistant rats. Singapore Med J. 2005; 46: 82–7.
  • El Mesallamy HO, El-Demerdash E, Hammad LN, El Magdoub HM. Effect of taurine supplementation on hyperhomocysteinemia and markers of oxidative stress in high fructose diet induced insulin resistance. Diabetol Metab Syndr. 2010; 2: 46.
  • Rahman MM, Park HM, Kim SJ, Go HK, Kim GB, Hong CU, Lee YU, Kim SZ, Kim JS, Kang HS. Taurine prevents hypertension and increases exercise capacity in rats with fructose-induced hypertension. Am J Hypertens. 2011 Feb 3. [Epub ahead of print].
  • Malone JI, Benford SA, Malone J Jr. Taurine prevents galactose-induced cataracts. J Diabetes Complications. 1993; 7: 44–8.
  • Huang JS, Chuang LY, Guh JY, Yang YL, Hsu MS. Effect of taurine on advanced glycation end products-induced hypertrophy in renal tubular epithelial cells. Toxicol Appl Pharmacol. 2008; 233: 220–6.
  • Huang JS, Chuang LY, Guh JY, Huang YJ. Effects of nitric oxide and antioxidants on advanced glycation end products-induced hypertrophic growth in human renal tubular cells. Toxicol Sci. 2009; 111: 109–19.
  • Nonaka H, Tsujino T, Watari Y, Emoto N, Yokoyama M. Taurine prevents the decrease in expression and secretion of extracellular superoxide dismutase induced by homocysteine: amelioration of homocysteine-induced endoplasmic reticulum stress by taurine. Circulation. 2001; 104: 1165–70.
  • Chang L, Xu J, Yu F, Zhao J, Tang X, Tang C. Taurine protected myocardial mitochondria injury induced by hyperhomocysteinemia in rats. Amino Acids. 2004; 27: 37–48.
  • Chang L, Xu JX, Zhao J, Pang YZ, Tang CS, Qi YF. Taurine antagonized oxidative stress injury induced by homocysteine in rat vascular smooth muscle cells. Acta Pharmacol Sin. 2004; 25: 341–6.
  • Chang L, Zhao J, Xu J, Jiang W, Tang CS, Qi YF. Effects of taurine and homocysteine on calcium homeostasis and hydrogen peroxide and superoxide anions in rat myocardial mitochondria. Clin Exp Pharmacol Physiol. 2004; 31: 237–43.
  • Li W, Li J, Deng B, Tian Y. [Effects of taurine on the heart defects in chick embryos which induced by homocysteine]. [Article in Chinese]. Wei Sheng Yan Jiu. 2004; 33: 183–5.
  • Balasubramanian T, Somasundaram M, Felix AJ. Taurine prevents ibuprofen-induced gastric mucosal lesions and influences endogenous antioxidant status of stomach in rats. ScientificWorldJournal. 2004; 4: 1046–54.
  • Son M, Kim HK, Kim WB, Yang J, Kim BK. Protective effect of taurine on indomethacin-induced gastric mucosal injury. Adv Exp Med Biol. 1996; 403: 147–55.
  • Motawi TK, Abd Elgawad HM, Shahin NN. Modulation of indomethacin-induced gastric injury by spermine and taurine in rats. J Biochem Mol Toxicol. 2007; 21: 280–8.
  • Izumi K, Nagata R, Motoya T, Yamashita J, Hirokane T, Nagata T, Satoh Y, Sawada Y, Ishibashi M, Yoshida H, et al. Preventive effect of taurine against acute paraquat intoxication in beagles. Jpn J Pharmacol. 1989; 50: 229–33.
  • Nagata T, Masaoka T, Akahori F. Protective effect of taurine against acute paraquat intoxication in rats. J Toxicol Sci. 1991; 16: 11–27.
  • Nakashima T, Taniko T, Kuriyama K. Therapeutic effect of taurine administration on carbon tetrachloride-induced hepatic injury. Jpn J Pharmacol. 1982; 32: 583–9.
  • Waterfield CJ, Mesquita M, Parnham P, Timbrell JA. Taurine protects against the cytotoxicity of hydrazine, 1,4-naphthoquinone and carbon tetrachloride in isolated rat hepatocytes. Biochem Pharmacol. 1993; 46: 589–95.
  • Waterfield CJ, Turton JA, Scales MD, Timbrell JA. Reduction of liver taurine in rats by beta-alanine treatment increases carbon tetrachloride toxicity. Toxicology. 1993; 77: 7–20.
  • Waterfield CJ, Mesquita M, Parnham P, Timbrell JA. Cytoprotective effects of taurine in isolated rat hepatocytes. Toxicol In Vitro. 1994; 8: 573–5.
  • Wu C, Miyagawa C, Kennedy DO, Yano Y, Otani S, Matsui-Yuasa I. Involvement of polyamines in the protection of taurine against the cytotoxicity of hydrazine or carbon tetrachloride in isolated rat hepatocytes. Chem Biol Interact. 1997; 103: 213–24.
  • Chen Y, Li S, Zhang X. [Taurine inhibits deposition of extracellular matrix in experimental liver fibrosis in rats]. [Article in Japanese]. Zhonghua Gan Zang Bing Za Zhi. 1999; 7: 165–7.
  • Wu C, Kennedy DO, Yano Y, Otani S, Matsui-Yuasa I. Thiols and polyamines in the cytoprotective effect of taurine on carbon tetrachloride-induced hepatotoxicity. J Biochem Mol Toxicol. 1999; 13: 71–6.
  • Vohra BP, Hui X. Taurine protects against carbon tetrachloride toxicity in the cultured neurons and in vivo. Arch Physiol Biochem. 2001; 109: 90–4.
  • Chen Y, Li S, Zhang X, Zhang Z, Xie W. [Amelioration of carbon tetrachloride-induced hepatic fibrosis in rats by treatment with Salvia miltiorrhiza and taurine]. [Article in Chinese]. Zhonghua Gan Zang Bing Za Zhi. 2002; 10: 148–9.
  • Dinçer S, Ozenirler S, Oz E, Akyol G, Ozoğul C. The protective effect of taurine pretreatment on carbon tetrachloride-induced hepatic damage-a light and electron microscopic study. Amino Acids. 2002; 22: 417–26.
  • Liang J, Zhang XL, Yang GY, Pang YS, Yuan HF, Liang JS, Huang RB. [Observation of the promotion effect taurine on hepatic stellate cell's apoptosis in rat hepatic fibrosis model]. [Article in Chinese]. Sichuan Da Xue Xue Bao Yi Xue Ban. 2005; 36: 365–7.
  • Miyazaki T, Karube M, Matsuzaki Y, Ikegami T, Doy M, Tanaka N, Bouscarel B. Taurine inhibits oxidative damage and prevents fibrosis in carbon tetrachloride-induced hepatic fibrosis. J Hepatol. 2005; 43: 117–25.
  • Tasci I, Mas MR, Vural SA, Deveci S, Comert B, Alcigir G, Mas N, Akay C, Bozdayi M, Yurdaydin C, Bozkaya H, Uzunalimoglu O, Isik AT, Said HM. Pegylated interferon-alpha plus taurine in treatment of rat liver fibrosis. World J Gastroenterol. 2007; 13: 3237–44.
  • Miyazaki T, Bouscarel B, Ikegami T, Honda A, Matsuzaki Y. The protective effect of taurine against hepatic damage in a model of liver disease and hepatic stellate cells. Adv Exp Med Biol. 2009; 643: 293–303.
  • Devamanoharan PS, Ali AH, Varma SD. Oxidative stress to rat lens in vitro: protection by taurine. Free Radic Res. 1998; 29: 189–95.
  • Mas MR, Isik AT, Yamanel L, Inal V, Tasci I, Deveci S, Mas N, Comert B, Akay C. Antioxidant treatment with taurine ameliorates chronic pancreatitis in an experimental rat model. Pancreas. 2006; 33: 77–81.
  • Giriş M, Depboylu B, Doğru-Abbasoğlu S, Erbil Y, Olgaç V, Aliş H, et al. Effect of taurine on oxidative stress and apoptosis-related protein expression in trinitrobenzene sulphonic acid-induced colitis. Clin Exp Immunol. 2008; 152: 102–10.
  • Mahalakshmi K, Pushpakiran G, Anuradha CV. Taurine prevents acrylonitrile-induced oxidative stress in rat brain. Pol J Pharmacol. 2003; 55: 1037–43.
  • Ebrahim AS, Babu E, Thirunavukkarasu C, Sakthisekaran D. Protective role of vitamin E, 2-deoxy-D-glucose, and taurine on perchloroethylene induced alterations in ATPases. Drug Chem Toxicol. 2001; 24: 429–37.
  • Seabra V, Timbrell JA. Modulation of taurine levels in the rat liver alters methylene dianiline hepatotoxicity. Toxicology. 1997; 122: 193–204.
  • Sinha M, Manna P, Sil PC. Taurine, a conditionally essential amino acid, ameliorates arsenic-induced cytotoxicity in murine hepatocytes. Toxicol In Vitro. 2007; 21: 1419–28.
  • Flora SJ, Chouhan S, Kannan GM, Mittal M, Swarnkar H. Combined administration of taurine and monoisoamyl DMSA protects arsenic induced oxidative injury in rats. Oxid Med Cell Longev. 2008; 1: 39–45.
  • Das J, Ghosh J, Manna P, Sinha M, Sil PC. Taurine protects rat testes against NaAsO(2)-induced oxidative stress and apoptosis via mitochondrial dependent and independent pathways. Toxicol Lett. 2009; 187: 201–10.
  • Das J, Ghosh J, Manna P, Sinha M, Sil PC. Arsenic-induced oxidative cerebral disorders: protection by taurine. Drug Chem Toxicol. 2009; 32: 93–102.
  • Ghosh J, Das J, Manna P, Sil PC. Taurine prevents arsenic-induced cardiac oxidative stress and apoptotic damage: role of NF-kappa B, p38 and JNK MAPK pathway. Toxicol Appl Pharmacol. 2009; 240: 73–87.
  • Li Z, Piao F, Liu S, Shen L, Sun N, Li B, Qu S. Preventive effects of taurine and vitamin C on renal DNA damage of mice exposed to arsenic. J Occup Health. 2009; 51: 169–72.
  • Roy A, Manna P, Sil PC. Prophylactic role of taurine on arsenic mediated oxidative renal dysfunction via MAPKs/ NF-kappaB and mitochondria dependent pathways. Free Radic Res. 2009; 43: 995–1007.
  • Das J, Ghosh J, Manna P, Sil PC. Protective role of taurine against arsenic-induced mitochondria-dependent hepatic apoptosis via the inhibition of PKCdelta-JNK pathway. PLoS One. 2010; 5: e12602.
  • Ma N, Sasoh M, Kawanishi S, Sugiura H, Piao F. Protection effect of taurine on nitrosative stress in the mice brain with chronic exposure to arsenic. J Biomed Sci. 2010; 17(Suppl 1): S7.
  • Hwang DF, Wang LC. Effect of taurine on toxicity of cadmium in rats. Toxicology. 2001; 167: 173–80.
  • Manna P, Sinha M, Sil PC. Amelioration of cadmium-induced cardiac impairment by taurine. Chem Biol Interact. 2008; 174: 88–97.
  • Manna P, Sinha M, Sil PC. Cadmium induced testicular pathophysiology: prophylactic role of taurine. Reprod Toxicol. 2008; 26: 282–91.
  • Sinha M, Manna P, Sil PC. Cadmium-induced neurological disorders: prophylactic role of taurine. J Appl Toxicol. 2008; 28: 974–86.
  • Sinha M, Manna P, Sil PC. Taurine protects the antioxidant defense system in the erythrocytes of cadmium treated mice. BMB Rep. 2008; 41: 657–63.
  • Kumar P, Prasad Y, Patra AK, Ranjan R, Swarup D, Patra RC, Pal S. Ascorbic acid, garlic extract and taurine alleviate cadmium-induced oxidative stress in freshwater catfish (Clarias batrachus). Sci Total Environ. 2009; 407: 5024–30.
  • Manna P, Sinha M, Sil PC. Taurine plays a beneficial role against cadmium-induced oxidative renal dysfunction. Amino Acids. 2009; 36: 417–28.
  • Sinha M, Manna P, Sil PC. Induction of necrosis in cadmium-induced hepatic oxidative stress and its prevention by the prophylactic properties of taurine. J Trace Elem Med Biol. 2009; 23: 300–13.
  • Jagadeesan G, Sankarsami Pillai S. Hepatoprotective effects of taurine against mercury induced toxicity in rats. J Environ Biol. 2007; 28: 753–6.
  • Neal R, Cooper K, Kellogg G, Gurer H, Ercal N. Effects of some sulfur-containing antioxidants on lead-exposed lenses. Free Radic Biol Med. 1999; 26: 239–43.
  • Hu JD, Gao QH, Yu DG, Xu XT. [The improvement of taurine in learning and memory ability of rats exposed to lead].[Article in Chinese]. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi. 2003; 21: 413–6.
  • Yang F, Li JS, Yan P, Liu YH, Wang DN. [Effect of taurine on NOS activity in hippocampus of rat exposed lead]. [Article in Chinese]. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi. 2004; 22: 203–6.
  • Zhu DM, Wang M, She JQ, Yu K, Ruan DY. Protection by a taurine supplemented diet from lead-induced deficits of long-term potentiation/depotentiation in dentate gyrus of rats in vivo. Neuroscience. 2005; 134: 215–24.
  • Yu SS, Wang M, Li XM, Chen WH, Chen JT, Wang HL, Ruan DY. Influences of different developmental periods of taurine supplements on synaptic plasticity in hippocampal CA1 area of rats following prenatal and perinatal lead exposure. BMC Dev Biol. 2007; 7: 51.
  • Fan G, Feng C, Li Y, Wang C, Yan J, Li W, Feng J, Shi X, Bi Y. Selection of nutrients for prevention or amelioration of lead-induced learning and memory impairment in rats. Ann Occup Hyg. 2009; 53: 341–51.
  • Hwang DF, Wang LC, Cheng HM. Effect of taurine on toxicity of copper in rats. Food Chem Toxicol. 1998; 36: 239–44.
  • Pasantes-Morales H, Wright CE, Gaull GE. Taurine protection of lymphoblastoid cells from iron-ascorbate induced damage. Biochem Pharmacol. 1985; 34: 2205–7.
  • Oudit GY, Trivieri MG, Khaper N, Husain T, Wilson GJ, Liu P, Sole MJ, Backx PH. Taurine supplementation reduces oxidative stress and improves cardiovascular function in an iron-overload murine model. Circulation. 2004; 109: 1877–85.
  • Otto-Duessel M, Aguilar M, Moats R, Wood JC. Antioxidant-mediated effects in a gerbil model of iron overload. Acta Haematol. 2007; 118: 193–9.
  • Schuller-Levis G, Quinn MR, Wright C, Park E. Taurine protects against oxidant-induced lung injury: possible mechanism(s) of action. Adv Exp Med Biol. 1994; 359: 31–9.
  • Qi B, Yamagami T, Naruse Y, Sokejima S, Kagamimori S. Effects of taurine on depletion of erythrocyte membrane Na-K ATPase activity due to ozone exposure or cholesterol enrichment. J Nutr Sci Vitaminol (Tokyo). 1995; 41: 627–34.
  • Schuller-Levis GB, Gordon RE, Park E, Pendino KJ, Laskin DL. Taurine protects rat bronchioles from acute ozone-induced lung inflammation and hyperplasia. Exp Lung Res. 1995; 21: 877–88.
  • Gordon RE, Park E, Laskin D, Schuller-Levis GB. Taurine protects rat bronchioles from acute ozone exposure: a freeze fracture and electron microscopic study. Exp Lung Res. 1998; 24: 659–74.
  • Rivas-Arancibia S, Dorado-Martínez C, Borgonio-Pérez G, Hiriart-Urdanivia M, Verdugo-Diaz L, Durán-Vázquez A, Colin-Baranque L, Avila-Costa MR. Effects of taurine on ozone-induced memory deficits and lipid peroxidation levels in brains of young, mature, and old rats. Environ Res. 2000; 82: 7–17.
  • Gordon RE, Shaked AA, Solano DF. Taurine protects hamster bronchioles from acute NO2-induced alterations. A histologic, ultrastructural, and freeze-fracture study. Am J Pathol. 1986; 125: 585–600.
  • Ogasawara M, Nakamura T, Koyama I, Nemoto M, Yoshida T. Reactivity of taurine with aldehydes and its physiological role. Chem Pharm Bull (Tokyo). 1993; 41: 2172–5.
  • Ogasawara M, Nakamura T, Koyama I, Nemoto M, Yoshida T. Reactivity of taurine with aldehydes and its physiological role. Adv Exp Med Biol. 1994; 359: 71–8.
  • Li G, Tang T, Peng M, He H, Yin D. Direct reaction of taurine with malondialdehyde: evidence for taurine as a scavenger of reactive carbonyl species. Redox Rep. 2010; 15: 268–74.
  • Marcinkiewicz J, Chain B, Nowak B, Grabowska A, Bryniarski K, Baran J. Antimicrobial and cytotoxic activity of hypochlorous acid: interactions with taurine and nitrite. Inflamm Res. 2000; 49: 280–9.
  • Marcinkiewicz J, Mak M, Bobek M, Biedroń R, Bialecka A, Koprowski M, Kontny E, Maśliński W. Is there a role of taurine bromamine in inflammation? Interactive effects with nitrite and hydrogen peroxide. Inflamm Res. 2005; 54: 42–9.
  • Marcinkiewicz J, Biedroń R, Bialecka A, Kasprowicz A, Mak M, Targosz M. Susceptibility of Propionibacterium acnes and Staphylococcus epidermidis to killing by MPO-halide system products. Implication for taurine bromamine as a new candidate for topical therapy in treating acne vulgaris. Arch Immunol Ther Exp (Warsz). 2006; 54: 61–8.
  • Kim C, Kim S. Taurine chloramine inhibits LPS-induced glucose uptake and glucose transporter 1 expression in RAW 264.7 macrophages. Adv Exp Med Biol. 2009; 643: 473–80.
  • Zulli A. Taurine in cardiovascular disease. Curr Opin Clin Nutr Metab Care. 2011; 14: 57–60.
  • Martinez-Losa M, Cortijo J, Piqueras L, Sanz MJ, Morcillo EJ. Taurine chloramine inhibits functional responses of human eosinophils in vitro. Clin Exp Allergy. 2009; 39: 537–46.
  • Kontny E, Rudnicka W, Chorazy-Massalska M, Marcinkiewicz J, Maśliński W. Taurine chloramine inhibits proliferation of rheumatoid arthritis synoviocytes by triggering a p53-dependent pathway. Inflamm Res. 2006; 55: 446–55.
  • Marcinkiewicz J, Kurnyta M, Biedroń R, Bobek M, Kontny E, Maśliński W. Anti-inflammatory effects of taurine derivatives (taurine chloramine, taurine bromamine, and taurolidine) are mediated by different mechanisms. Adv Exp Med Biol. 2006; 583: 481–92.
  • Muz B, Kontny E, Marcinkiewicz J, Maśliński W. Heme oxygenase-1 participates in the anti-inflammatory activity of taurine chloramine. Amino Acids. 2008; 35: 397–402.
  • Kanayama A, Inoue J, Sugita-Konishi Y, Shimizu M, Miyamoto Y. Oxidation of Ikappa Balpha at methionine 45 is one cause of taurine chloramine-induced inhibition of NF-kappa B activation. J Biol Chem. 2002; 277: 24049–56.
  • Ogino T, Hosako M, Hiramatsu K, Omori M, Ozaki M, Okada S. Oxidative modification of IkappaB by monochloramine inhibits tumor necrosis factor alpha-induced NF-kappaB activation. Biochim Biophys Acta. 2005; 1746: 135–42.
  • Midwinter RG, Cheah FC, Moskovitz J, Vissers MC, Winterbourn CC. IkappaB is a sensitive target for oxidation by cell-permeable chloramines: inhibition of NF-kappaB activity by glycine chloramine through methionine oxidation. Biochem J. 2006; 396: 71–8.
  • Tokunaga S, Kanayama A, Miyamoto Y. Modification of IkappaBalpha by taurine bromamine inhibits tumor necrosis factor alpha-induced NF-kappaB activation. Inflamm Res. 2007; 56: 479–86.
  • Hart LA, Krishnan VL, Adcock IM, Barnes PJ, Chung KF. Activation and localization of transcription factor, nuclear factor-kappaB, in asthma. Am J Respir Crit Care Med. 1998; 158: 1585–92.
  • Dong G, Chen Z, Kato T, Van Waes C. The host environment promotes the constitutive activation of nuclear factor-kappaB and proinflammatory cytokine expression during metastatic tumor progression of murine squamous cell carcinoma. Cancer Res. 1999; 59: 3495–504.
  • Gan HT, Chen YQ, Ouyang Q. Sulfasalazine inhibits activation of nuclear factor-kappaB in patients with ulcerative colitis. J Gastroenterol Hepatol. 2005; 20: 1016–24.
  • Kramer JH, Chovan JP, Schaffer SW. Effect of taurine on calcium paradox and ischemic heart failure. Am J Physiol. 1981; 240: H238–46.
  • Pukhova TM, Zhuchkova NI. [The effect of taurine on the density of adrenergic nerve endings and the recovery of cardiac function after ischemia]. [Article in Russian]. Biull Eksp Biol Med. 1991; 111: 241–4.
  • Milei J, Ferreira R, Llesuy S, Forcada P, Covarrubias J, Boveris A. Reduction of reperfusion injury with preoperative rapid intravenous infusion of taurine during myocardial revascularization. Am Heart J. 1992; 123: 339–45.
  • Kapel'ko VI, Pisarenko OI, Pukhova TM, Lakomkin VL, Solomatina ES, Studneva IM, Novikova NA. [Modifications to the solution for reperfusion of the ischemic heart]. [Article in Russian]. Kardiologiia. 1993; 33: 71–4, 7.
  • Raschke P, Massoudy P, Becker BF. Taurine protects the heart from neutrophil-induced reperfusion injury. Free Radic Biol Med. 1995; 19: 461–71.
  • Chahine R, Feng J. Protective effects of taurine against reperfusion-induced arrhythmias in isolated ischemic rat heart. Arzneimittelforschung. 1998; 48: 360–4.
  • Satoh H, Sperelakis N. Review of some actions of taurine on ion channels of cardiac muscle cells and others. Gen Pharmacol. 1998; 30: 451–63.
  • Oz E, Erbaş D, Gelir E, Aricioğlu A. Taurine and calcium interaction in protection of myocardium exposed to ischemic reperfusion injury. Gen Pharmacol. 1999; 33: 137–41.
  • Takahashi K, Ohyabu Y, Schaffer SW, Azuma J. Taurine prevents ischemia damage in cultured neonatal rat cardiomyocytes. Adv Exp Med Biol. 2000; 483: 109–16.
  • Takahashi K, Ohyabu Y, Takahashi K, Solodushko V, Takatani T, Itoh T, Schaffer SW, Azuma J. Taurine renders the cell resistant to ischemia-induced injury in cultured neonatal rat cardiomyocytes. J Cardiovasc Pharmacol. 2003; 41: 726–33.
  • Hanna J, Chahine R, Aftimos G, Nader M, Mounayar A, Esseily F, Chamat S. Protective effect of taurine against free radicals damage in the rat myocardium. Exp Toxicol Pathol. 2004; 56: 189–94.
  • Kingston R, Kelly CJ, Murray P. The therapeutic role of taurine in ischaemia-reperfusion injury. Curr Pharm Des. 2004; 10: 2401–10.
  • Li AY, Ji ES, Zhao SM, Ma ZH, Li Q. [Effects of taurine on rabbit cardiomyocyte apoptosis during ischemia/reperfusion injury]. [Article in Chinese]. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 2004; 20: 224–7.
  • Takatani T, Takahashi K, Uozumi Y, Matsuda T, Ito T, Schaffer SW, Fujio Y, Azuma J. Taurine prevents the ischemia-induced apoptosis in cultured neonatal rat cardiomyocytes through Akt/caspase-9 pathway. Biochem Biophys Res Commun. 2004; 316: 484–9.
  • Takatani T, Takahashi K, Uozumi Y, Shikata E, Yamamoto Y, Ito T, Matsuda T, Schaffer SW, Fujio Y, Azuma J. Taurine inhibits apoptosis by preventing formation of the Apaf-1/caspase-9 apoptosome. Am J Physiol Cell Physiol. 2004; 287: C949–53.
  • Oriyanhan W, Yamazaki K, Miwa S, Takaba K, Ikeda T, Komeda M. Taurine prevents myocardial ischemia/reperfusion-induced oxidative stress and apoptosis in prolonged hypothermic rat heart preservation. Heart Vessels. 2005; 20: 278–85.
  • Oriyanhan W, Miyamoto TA, Yamazaki K, Miwa S, Takaba K, Ikeda T, Komeda M. Regionally perfused taurine. Part I. Minimizes lactic acidosis and preserves CKMB and myocardial contractility after ischemia/reperfusion. Adv Exp Med Biol. 2006; 583: 271–88.
  • Takahashi K, Takatani T, Uozumi Y, Ito T, Matsuda T, Fujio Y, Schaffer SW, Azuma J. Molecular mechanisms of cardioprotection by taurine on ischemia-induced apoptosis in cultured cardiomyocytes. Adv Exp Med Biol. 2006; 583: 257–63.
  • Ueno T, Iguro Y, Yotsumoto G, Fukumoto Y, Nakamura K, Miyamoto TA, Sakata R. Taurine at early reperfusion significantly reduces myocardial damage and preserves cardiac function in the isolated rat heart. Resuscitation. 2007; 73: 287–95.
  • Briet F, Keith M, Leong-Poi H, Kadakia A, Aba-Alkhail K, Giliberto JP, Stewart D, Errett L, David Mazer C. Triple nutrient supplementation improves survival, infarct size and cardiac function following myocardial infarction in rats. Nutr Metab Cardiovasc Dis. 2008; 18: 691–9.
  • Doddakula KK, Neary PM, Wang JH, Sookhai S, O'Donnell A, Aherne T, Bouchier-Hayes DJ, Redmond HP. The antiendotoxin agent taurolidine potentially reduces ischemia/reperfusion injury through its metabolite taurine. Surgery. 2010; 148: 567–72.
  • Kulthinee S, Wyss JM, Jirakulsomchok D, Roysommuti S. High sugar intake exacerbates cardiac reperfusion injury in perinatal taurine depleted adult rats. J Biomed Sci. 2010; 17(Suppl 1): S22.
  • McLaughlin R, Bowler D, Kelly CJ, Kay E, Bouchier-Hayes D. Taurine protects against early and late skeletal muscle dysfunction secondary to ischaemia reperfusion injury. Eur J Surg. 2000; 166: 375–9.
  • Kingston R, Kearns S, Kelly C, Murray P. Effects of systemic and regional taurine on skeletal muscle function following ischaemia-reperfusion injury. J Orthop Res. 2005; 23: 310–4.
  • Wang JX, Li Y, Zhang LK, Zhao J, Pang YZ, Tang CS, Zhang J. Taurine inhibits ischemia/reperfusion-induced compartment syndrome in rabbits. Acta Pharmacol Sin. 2005; 26: 821–7.
  • Zhang N, Zhang LY, Wang YH, Dong SY, Kong XY, Zhao LJ. [Effects of taurine on TNF-alpha and NF-kappaB expression of liver injury after limbs ischemia/reperfusion in rats]. [Article in Chinese]. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 2009; 25: 18–21.
  • Akdemir O, Hede Y, Zhang F, Lineaweaver WC, Arslan Z, Songur E. Effects of taurine on reperfusion injury. J Plast Reconstr Aesthet Surg. 2011 Jan 21. [Epub ahead of print].
  • Chen YX. [Protective action of taurine on ischemia-reperfusion liver injury in rats and its mechanism]. [Article in Chinese]. Zhonghua Yi Xue Za Zhi. 1993; 73(276–9): 318–9.
  • Minor T, Yamaguchi T, Isselhard W. Effects of taurine on liver preservation in UW solution with consecutive ischemic rewarming in the isolated perfused rat liver. Transpl Int. 1995; 8: 174–9.
  • Minor T, Yamaguchi T, Klauke H, Wingenfeld P, Michalk D, Isselhard W. Taurine reduces experimental liver injury after cold ischemic preservation and a period of rewarming prior to reperfusion. Adv Exp Med Biol. 1996; 403: 157–61.
  • Wettstein M, Häussinger D. Cytoprotection by the osmolytes betaine and taurine in ischemia-reoxygenation injury in the perfused rat liver. Hepatology. 1997; 26: 1560–6.
  • Wettstein M, Häussinger D. Taurine attenuates cold ischemia-reoxygenation injury in rat liver. Transplantation. 2000; 69: 2290–6.
  • Lauschke H, Kötting M, Akbar S, Minor T. Use of taurine as antioxidant in resuscitating livers from non-heart-beating donors by gaseous oxygen persufflation. J Invest Surg. 2003; 16: 7–11.
  • Schemmer P, Liang R, Kincius M, Flechtenmacher C, Bunzendahl H, Gutt CN, Mehrabi A, Gebhard MM, Büchler MW, Kraus TW. Taurine improves graft survival after experimental liver transplantation. Liver Transpl. 2005; 11: 950–9.
  • Tong L, Li J, Qiao H, Jiang H, Meng F, Sun X. Taurine protects against ischemia-reperfusion injury in rabbit livers. Transplant Proc. 2006; 38: 1575–9.
  • Kincius M, Liang R, Nickkholgh A, Hoffmann K, Flechtenmacher C, Ryschich E, Gutt CN, Gebhard MM, Schmidt J, Büchler MW, Schemmer P. Taurine protects from liver injury after warm ischemia in rats: the role of Kupffer cells. Eur Surg Res. 2007; 39: 275–83.
  • Schindler G, Kincius M, Liang R, Backhaus J, Zorn M, Flechtenmacher C, Gebhard MM, Büchler MW, Schemmer P. Fundamental efforts toward the development of a therapeutic cocktail with a manifold ameliorative effect on hepatic ischemia/reperfusion injury. Microcirculation. 2009; 16: 593–602.
  • Bruns H, Watanpour I, Gebhard MM, Flechtenmacher C, Galli U, Schulze-Bergkamen H, Zorn M, Büchler MW, Schemmer P. Glycine and taurine equally prevent fatty livers from Kupffer cell dependent injury: an in vivo microscopy study. Microcirculation. 2010 Dec 22. 10.3402/mehd.v23i0.14787. [Epub ahead of print].
  • Zhang F, Mao Y, Qiao H, Jiang H, Zhao H, Chen X, Tong L, Sun X. Protective effects of taurine against endotoxin-induced acute liver injury after hepatic ischemia reperfusion. Amino Acids. 2010; 38: 237–45.
  • Michalk DV, Hoffmann B, Minor T. Taurine reduces renal ischemia/reperfusion injury in the rat. Adv Exp Med Biol. 2003; 526: 49–56.
  • Guz G, Oz E, Lortlar N, Ulusu NN, Nurlu N, Demirogullari B, Omeroglu S, Sert S, Karasu C. The effect of taurine on renal ischemia/reperfusion injury. Amino Acids. 2007; 32: 405–11.
  • Guan X, Dei-Anane G, Liang R, Gross ML, Nickkholgh A, Kern M, Ludwig J, Zeier M, Büchler MW, Schmidt J, Schemmer P. Donor preconditioning with taurine protects kidney grafts from injury after experimental transplantation. J Surg Res. 2008; 146: 127–34.
  • Wei SM, Yan ZZ, Zhou J. Beneficial effect of taurine on testicular ischemia-reperfusion injury in rats. Urology. 2007; 70: 1237–42.
  • Namazi H. Novel molecular mechanism to account for action of taurine against testicular ischemia-reperfusion injury. Urology. 2008; 72: 465–6.
  • Wei SM, Yan ZZ, Zhou J. Taurine reduces testicular ischemia/reperfusion-induced neutrophil recruitment to testis probably by downregulation of pro-inflammatory cytokines and E-selectin. Urology. 2008; 72: 464–5.
  • Zhang F, Tong L, Qiao H, Dong X, Qiao G, Jiang H, Sun X. Taurine attenuates multiple organ injury induced by intestinal ischemia reperfusion in rats. J Surg Res. 2008; 149: 101–9.
  • Pessina F, Matteucci G, Esposito L, Gorelli B, Valoti M, Sgaragli G. Protection of intrinsic nerves of guinea-pig detrusor strips against anoxia/glucopenia and reperfusion injury by taurine. Adv Exp Med Biol. 2000; 483: 325–33.
  • Oz E, Sivrikoz MC, Halit V, Altunkaya A, Take G. The role of taurine added to pulmonary reperfusion solutions in isolated guinea pig lungs. Amino Acids. 2002; 22: 391–403.
  • Guo J, Li R, Zhao P, Cheng J. Effect of taurine in combination with electroacupuncture on neuronal damage following transient focal cerebral ischemia in rats. Acupunct Electrother Res. 2002; 27: 129–36.
  • Li M, Gao GD, Zheng J, Sun LZ, Long C. [Effects of taurine on early changes of excitatory amino acids in rabbit brain due to deep hypothermic circulatory arrest]. [Article in Chinese]. Zhonghua Wai Ke Za Zhi. 2005; 43: 362–5.
  • Molchanova SM, Oja SS, Saransaari P.2006; 1099: 64–72.
  • Wang GH, Jiang ZL, Fan XJ, Zhang L, Li X, Ke KF. Neuroprotective effect of taurine against focal cerebral ischemia in rats possibly mediated by activation of both GABAA and glycine receptors. Neuropharmacology. 2007; 52: 1199–209.
  • Sun M, Xu C. Neuroprotective mechanism of taurine due to up-regulating calpastatin and down-regulating calpain and caspase-3 during focal cerebral ischemia. Cell Mol Neurobiol. 2008; 28: 593–611.
  • Taranukhin AG, Taranukhina EY, Saransaari P, Djatchkova IM, Pelto-Huikko M, Oja SS. Taurine reduces caspase-8 and caspase-9 expression induced by ischemia in the mouse hypothalamic nuclei. Amino Acids. 2008; 34: 169–74.
  • Ricci L, Valoti M, Sgaragli G, Frosini M. Protection by taurine of rat brain cortical slices against oxygen glucose deprivation- and reoxygenation-induced damage. Eur J Pharmacol. 2009; 621: 26–32.
  • Sun M, Gu Y, Zhao Y, Xu C. Protective functions of taurine against experimental stroke through depressing mitochondria-mediated cell death in rats. Amino Acids. 2010 Sep 23. [Epub ahead of print].
  • Sun M, Zhao Y, Gu Y, Xu C. Anti-inflammatory mechanism of taurine against ischemic stroke is related to down-regulation of PARP and NF-κB. Amino Acids. 2011 Mar 16. [Epub ahead of print].
  • Chan PH. Oxygen radicals in focal cerebral ischemia. Brain Pathol. 1994; 4: 59–65.
  • Shi H, Liu KJ. Cerebral tissue oxygenation and oxidative brain injury during ischemia and reperfusion. Front Biosci. 2007; 12: 1318–28.
  • Zhang W, Wang M, Xie HY, Zhou L, Meng XQ, Shi J, Zheng S. Role of reactive oxygen species in mediating hepatic ischemia-reperfusion injury and its therapeutic applications in liver transplantation. Transplant Proc. 2007; 39: 1332–7.
  • Bhogal RH, Curbishley SM, Weston CJ, Adams DH, Afford SC.2010; 16: 1303–13.
  • Kahles T, Kohnen A, Heumueller S, Rappert A, Bechmann I, Liebner S, Wittko IM, Neumann-Haefelin T, Steinmetz H, Schroeder K, Brandes RP. NADPH oxidase Nox1 contributes to ischemic injury in experimental stroke in mice. Neurobiol Dis. 2010; 40: 185–92.
  • Li RC, Guo SZ, Lee SK, Gozal D. Neuroglobin protects neurons against oxidative stress in global ischemia. J Cereb Blood Flow Metab. 2010; 30: 1874–82.
  • Thu VT, Kim HK, Ha SH, Yoo JY, Park WS, Kim N, Oh GT, Han J. Glutathione peroxidase 1 protects mitochondria against hypoxia/reoxygenation damage in mouse hearts. Pflugers Arch. 2010; 460: 55–68.
  • Pasdois P, Parker JC, Griffiths EJ, Halestrap AP. The role of oxidized cytochrome c in regulating mitochondrial reactive oxygen species production and its perturbation in ischaemia. Biochem J. 2011 Mar 17. [Epub ahead of print].
  • Quarrie R, Cramer BM, Lee DS, Steinbaugh GE, Erdahl W, Pfeiffer DR, Zweier JL, Crestanello JA. Ischemic preconditioning decreases mitochondrial proton leak and reactive oxygen species production in the postischemic heart. J Surg Res. 2011; 165: 5–14.
  • Sandin A, Dagnell M, Gonon A, Pernow J, Stangl V, Aspenström P, Kappert K, Ostman A. Hypoxia followed by re-oxygenation induces oxidation of tyrosine phosphatases. Cell Signal. 2011; 23: 820–6.
  • Krasova EI, Nastashenko TA, Onishchenko NA, Seı˘fulla RD, Kuvaev AE. [Effect of alpha-tocopherol acetate and sodium selenite on the change in ATP content and the RNA synthesis rate in the ischemic myocardium]. [Article in Russian]. Farmakol Toksikol. 1979; 42: 251–4.
  • Litvitskiı˘ PF, Kogan AKh, Kudrin AN, Luk'ianova LO. [Pathogenetic role of lipid peroxidation and the protective role of sodium selenite in ischemia and myocardial reperfusion]. [Article in Russian]. Biull Eksp Biol Med. 1981; 91: 271–4.
  • Nath KA, Paller MS. Dietary deficiency of antioxidants exacerbates ischemic injury in the rat kidney. Kidney Int. 1990; 38: 1109–17.
  • Chiba Y, Muraoka R, Noguchi H, Hiramatsu Y, Kimura T, Ihaya A, Morioka K. [Significance of selenium deficiency on myocardial protection of the mature and immature rat hearts]. [Article in Japanese]. Nippon Kyobu Geka Gakkai Zasshi. 1991; 39: 1882–7.
  • Poltronieri R, Cevese A, Sbarbati A. Protective effect of selenium in cardiac ischemia and reperfusion. Cardioscience. 1992; 3: 155–60.
  • Soncul H, Kaptanoğlu M, Oz E, Halit V, Bilgehan A, Cayci B, Gökgöz L, Türkozan N, Ersöz A. The role of selenium added to pulmonary preservation solutions in isolated guinea pig lungs. J Thorac Cardiovasc Surg. 1994; 108: 922–7.
  • Soncul H, Tatlican O, Halit V, Oz E, Sinci V, Salman E, Gökgöz L, Türközkan N, Ersöz A. The effect of selenium added cardioplegia in guinea pigs. Gen Pharmacol. 1994; 25: 1493–7.
  • Erbas D, Soncul H, Turkozkan N, Arioioglu A, Muftugoglu S, Ersoz A. Effect of selenium on ischemic and reperfusion injury in isolated guinea pig lungs. Gen Pharmacol. 1995; 26: 1669–72.
  • Boucher F, Coudray C, Tirard V, Barandier C, Tresallet N, Favier A, de Leiris J. Oral selenium supplementation in rats reduces cardiac toxicity of adriamycin during ischemia and reperfusion. Nutrition. 1995; 11((5 Suppl)): 708–11.
  • Pucheu S, Coudray C, Tresallet N, Favier A, de Leiris J. Effect of dietary antioxidant trace element supply on cardiac tolerance to ischemia-reperfusion in the rat. J Mol Cell Cardiol. 1995; 27: 2303–14.
  • Sinci V, Gunaydin S, Kalaycioglu S, Soncul H, Gokgoz L, Oz E. Effects of selenium enriched reperfusion solutions on isolated guinea pig hearts. Keio J Med. 1998; 47: 219–22.
  • Tanguy S, Boucher F, Besse S, Ducros V, Favier A, de Leiris J. Trace elements and cardioprotection: increasing endogenous glutathione peroxidase activity by oral selenium supplementation in rats limits reperfusion-induced arrhythmias. J Trace Elem Med Biol. 1998; 12: 28–38.
  • Huang Y, Bai H, Zhang Z. [Mechanism of selenium protecting against free radical damages during myocardial ischemia/reperfusion in rats]. [Article in Chinese]. Zhonghua Yi Xue Za Zhi. 1999; 79: 852–6.
  • Huang Y, Liu Y, Zhang Z. [Mechanism of selenium defending against free radical damages during myocardial ischemia/reperfusion in human]. [Article in Chinese]. Zhonghua Yi Xue Za Zhi. 1999; 79: 731–4.
  • Oztürk C, Avlan D, Cinel I, Cinel L, Unlü A, Camdeviren H, Atik U, Oral U. Selenium pretreatment prevents bacterial translocation in rat intestinal ischemia/reperfusion model. Pharmacol Res. 2002; 46: 171–5.
  • Gupta R, Singh M, Sharma A. Neuroprotective effect of antioxidants on ischaemia and reperfusion-induced cerebral injury. Pharmacol Res. 2003; 48: 209–15.
  • Tanguy S, Toufektsian MC, Besse S, Ducros V, De Leiris J, Boucher F. Dietary selenium intake affects cardiac susceptibility to ischaemia/reperfusion in male senescent rats. Age Ageing. 2003; 32: 273–8.
  • Treska V, Kuntscher V, Molácek J, Kobr J, Racek J, Trefil L. Can ischemia-reperfusion syndrome in transplanted kidneys procured from non-heart-beating donors be influenced by adding selenium into the reperfusion solution? An experimental study. Transplant Proc. 2003; 35: 3125–7.
  • Treska V, Kuntscher V, Molácek J, Kobr J, Racek J, Trefil L. Can the ischemia-reperfusion syndrome in transplanted kidneys procured from non-heart-beating donors be influenced by adding selenium into the reperfusion solution? An experimental study. Transplant Proc. 2003; 35: 1584–6.
  • Ansari MA, Ahmad AS, Ahmad M, Salim S, Yousuf S, Ishrat T, Islam F. Selenium protects cerebral ischemia in rat brain mitochondria. Biol Trace Elem Res. 2004; 101: 73–86.
  • Tanguy S, Morel S, Berthonneche C, Toufektsian MC, de Lorgeril M, Ducros V, Tosaki A, de Leiris J, Boucher F. Preischemic selenium status as a major determinant of myocardial infarct size in vivo in rats. Antioxid Redox Signal. 2004; 6: 792–6.
  • Venardos K, Harrison G, Headrick J, Perkins A. Effects of dietary selenium on glutathione peroxidase and thioredoxin reductase activity and recovery from cardiac ischemia-reperfusion. J Trace Elem Med Biol. 2004; 18: 81.
  • Venardos K, Harrison G, Headrick J, Perkins A. Selenium supplementation and ischemia-reperfusion injury in rats. Redox Rep. 2004; 9: 317–20.
  • Avlan D, Erdouğan K, Cimen B, Düşmez Apa D, Cinel I, Aksöyek S. The protective effect of selenium on ipsilateral and contralateral testes in testicular reperfusion injury. Pediatr Surg Int. 2005; 21: 274–8.
  • Rakotovao A, Tanguy S, Toufektsian MC, Berthonneche C, Ducros V, Tosaki A, de Leiris J, Boucher F. Selenium status as determinant of connexin-43 dephosphorylation in ex vivo ischemic/reperfused rat myocardium. J Trace Elem Med Biol. 2005; 19: 43–7.
  • Turan B, Saini HK, Zhang M, Prajapati D, Elimban V, Dhalla NS. Selenium improves cardiac function by attenuating the activation of NF-kappaB due to ischemia-reperfusion injury. Antioxid Redox Signal. 2005; 7: 1388–97.
  • Venardos K, Ashton K, Headrick J, Perkins A. Effects of dietary selenium on post-ischemic expression of antioxidant mRNA. Mol Cell Biochem. 2005; 270: 131–8.
  • Lymbury R, Venardos K, Perkins AV. Effect of sodium selenite-enriched reperfusion solutions on rat cardiac ischemia reperfusion injury. Biol Trace Elem Res. 2006; 114: 197–206.
  • Ostadalova I, Vobecky M, Chvojkova Z, Mikova D, Hampl V, Wilhelm J, Ostadal B. Selenium protects the immature rat heart against ischemia/reperfusion injury. Mol Cell Biochem. 2007; 300: 259–67.
  • Venardos KM, Perkins A, Headrick J, Kaye DM. Myocardial ischemia-reperfusion injury, antioxidant enzyme systems, and selenium: a review. Curr Med Chem. 2007;14:1539-49. Erratum in: Curr Med Chem. 2007;14:2944. Perkins, Anthony [added]; Headrick, John [added].
  • Yousuf S, Atif F, Ahmad M, Hoda MN, Khan MB, Ishrat T, Islam F. Selenium plays a modulatory role against cerebral ischemia-induced neuronal damage in rat hippocampus. Brain Res. 2007; 1147: 218–25.
  • Erbil G, Ozbal S, Sonmez U, Pekcetin C, Tugyan K, Bagriyanik A, Ozogul C. Neuroprotective effects of selenium and Ginkgo biloba extract (EGb761) against ischemia and reperfusion injury in rat brain. Neurosciences (Riyadh). 2008; 13: 233–8.
  • Ozbal S, Erbil G, Koçdor H, Tuğyan K, Pekçetin C, Ozoğul C. The effects of selenium against cerebral ischemia-reperfusion injury in rats. Neurosci Lett. 2008; 438: 265–9.
  • Zapletal C, Heyne S, Breitkreutz R, Gebhard MM, Golling M. The influence of selenium substitution on microcirculation and glutathione metabolism after warm liver ischemia/reperfusion in a rat model. Microvasc Res. 2008; 76: 104–9.
  • Tanguy S, Rakotovao A, Jouan MG, Ghezzi C, de Leiris J, Boucher F. Dietary selenium intake influences Cx43 dephosphorylation, TNF-α expression and cardiac remodeling after reperfused infarction. Mol Nutr Food Res. 2010 Dec 15. [Epub ahead of print].
  • Wang GS, Geng DQ, Wang YW, Chen XD, Yang TH, Chang CH. [Protective effect of Na2SeO3 against cerebral ischemia-reperfusion injury to the hippocampal neurons in rats]. [Article in Chinese]. Nan Fang Yi Ke Da Xue Xue Bao. 2010; 30: 2336–9.
  • Blaustein A, Deneke SM, Stolz RI, Baxter D, Healey N, Fanburg BL. Myocardial glutathione depletion impairs recovery after short periods of ischemia. Circulation. 1989; 80: 1449–57.
  • Singh A, Lee KJ, Lee CY, Goldfarb RD, Tsan MF. Relation between myocardial glutathione content and extent of ischemia-reperfusion injury. Circulation. 1989; 80: 1795–804.
  • Nishinaka Y, Kitahara S, Sugiyama S, Yokota M, Saito H, Ozawa T. The cardioprotective effect of gamma-glutamylcysteine ethyl ester during coronary reperfusion in canine hearts. Br J Pharmacol. 1991; 104: 805–10.
  • Werns SW, Fantone JC, Ventura A, Lucchesi BR. Myocardial glutathione depletion impairs recovery of isolated blood-perfused hearts after global ischaemia. J Mol Cell Cardiol. 1992; 24: 1215–20.
  • Guarnieri C, Turinetto B, Colì G, Muscari C, Cattabriga I, Vaona I, Finelli C, Pigini F, Caldarera CM. Effect of glutathione monoethyl ester on glutathione level and cardiac energetics in reperfused pig heart. Res Commun Chem Pathol Pharmacol. 1993; 81: 33–44.
  • Nakano H, Boudjema K, Alexandre E, Imbs P, Chenard MP, Wolf P, Cinqualbre J, Jaeck D. Protective effects of N-acetylcysteine on hypothermic ischemia-reperfusion injury of rat liver. Hepatology. 1995; 22: 539–45.
  • Nakano H, Nagasaki H, Barama A, Boudjema K, Jaeck D, Kumada K, Tatsuno M, Baek Y, Kitamura N, Suzuki T, Yamaguchi M. The effects of N-acetylcysteine and anti-intercellular adhesion molecule-1 monoclonal antibody against ischemia-reperfusion injury of the rat steatotic liver produced by a choline-methionine-deficient diet. Hepatology. 1997; 26: 670–8.
  • Nakano H, Nagasaki H, Yoshida K, Kigawa G, Fujiwara Y, Kitamura N, Kuzume M, Takeuchi S, Sasaki J, Shimura H, Yamaguchi M, Kumada K. N-acetylcysteine and anti-ICAM-1 monoclonal antibody reduce ischemia-reperfusion injury of the steatotic rat liver. Transplant Proc. 1998; 30: 3763.
  • Bilzer M, Paumgartner G, Gerbes AL. Glutathione protects the rat liver against reperfusion injury after hypothermic preservation. Gastroenterology. 1999; 117: 200–10.
  • Grattagliano I, Vendemiale G, Lauterburg BH. Reperfusion injury of the liver: role of mitochondria and protection by glutathione ester. J Surg Res. 1999; 86: 2–8.
  • Paterson PG, Juurlink BH. Nutritional regulation of glutathione in stroke. Neurotox Res. 1999; 1: 99–112.
  • Cheung PY, Wang W, Schulz R. Glutathione protects against myocardial ischemia-reperfusion injury by detoxifying peroxynitrite. J Mol Cell Cardiol. 2000; 32: 1669–78.
  • Leichtweis S, Ji LL. Glutathione deficiency intensifies ischaemia-reperfusion induced cardiac dysfunction and oxidative stress. Acta Physiol Scand. 2001; 172: 1–10.
  • Leichtweis S, Leeuwenburgh C, Bejma J, Ji LL. Aged rat hearts are not more susceptible to ischemia-reperfusion injury in vivo: role of glutathione. Mech Ageing Dev. 2001; 122: 503–18.
  • Ramires PR, Ji LL. Glutathione supplementation and training increases myocardial resistance to ischemia-reperfusion in vivo. Am J Physiol Heart Circ Physiol. 2001; 281: H679–88.
  • Bilzer M, Baron A, Schauer R, Steib C, Ebensberger S, Gerbes AL. Glutathione treatment protects the rat liver against injury after warm ischemia and Kupffer cell activation. Digestion. 2002; 66: 49–57.
  • Bobyn PJ, Franklin JL, Wall CM, Thornhill JA, Juurlink BH, Paterson PG. The effects of dietary sulfur amino acid deficiency on rat brain glutathione concentration and neural damage in global hemispheric hypoxia-ischemia. Nutr Neurosci. 2002; 5: 407–16.
  • Shen WH, Zhang CY, Zhang GY. Antioxidants attenuate reperfusion injury after global brain ischemia through inhibiting nuclear factor-kappa B activity in rats. Acta Pharmacol Sin. 2003; 24: 1125–30.
  • Anderson MF, Nilsson M, Eriksson PS, Sims NR. Glutathione monoethyl ester provides neuroprotection in a rat model of stroke. Neurosci Lett. 2004; 354: 163–5.
  • Anderson MF, Nilsson M, Sims NR. Glutathione monoethylester prevents mitochondrial glutathione depletion during focal cerebral ischemia. Neurochem Int. 2004; 44: 153–9.
  • Kupatt C, Hinkel R, Horstkotte J, Deiss M, von Brühl ML, Bilzer M, Boekstegers P. Selective retroinfusion of GSH and cariporide attenuates myocardial ischemia-reperfusion injury in a preclinical pig model. Cardiovasc Res. 2004; 61: 530–7.
  • Schauer RJ, Gerbes AL, Vonier D, Meissner H, Michl P, Leiderer R, Schildberg FW, Messmer K, Bilzer M. Glutathione protects the rat liver against reperfusion injury after prolonged warm ischemia. Ann Surg. 2004; 239: 220–31.
  • Schauer RJ, Kalmuk S, Gerbes AL, Leiderer R, Meissner H, Schildberg FW, Messmer K, Bilzer M. Intravenous administration of glutathione protects parenchymal and non-parenchymal liver cells against reperfusion injury following rat liver transplantation. World J Gastroenterol. 2004; 10: 864–70.
  • Sims NR, Nilsson M, Muyderman H. Mitochondrial glutathione: a modulator of brain cell death. J Bioenerg Biomembr. 2004; 36: 329–33.
  • Pratschke S, Angele MK, Grützner U, Tufman A, Bilzer M, Loehe F, Jauch KW, Schauer RJ. GSH attenuates organ injury and improves function after transplantation of fatty livers. Eur Surg Res. 2010; 45: 13–9.
  • Ye S, Dong J, Han B. Protective effect of reduced glutathione and venous systemic oxygen persufflation on rat steatotic graft following liver transplantation. J Surg Res. 2010; 158: 138–46.
  • Rusakov VV, Dolgikh VT. [Reperfusion injury of myocardial biomembranes after acute fatal hemorrhage and their correction with carnosine]. [Article in Russian]. Biokhimiia. 1992; 57: 1393–7.
  • Alabovsky VV, Boldyrev AA, Vinokurov AA, Shchavratsky VKh. Effect of histidine-containing dipeptides on isolated heart under ischemia/reperfusion. Biochemistry (Mosc). 1997; 62: 77–87.
  • Fujii T, Takaoka M, Muraoka T, Kurata H, Tsuruoka N, Ono H, Kiso Y, Tanaka T, Matsumura Y. Preventive effect of L-carnosine on ischemia/reperfusion-induced acute renal failure in rats. Eur J Pharmacol. 2003; 474: 261–7.
  • Dobrota D, Fedorova T, Stvolinsky S, Babusikova E, Likavcanova K, Drgova A, Strapkova A, Boldyrev A. Carnosine protects the brain of rats and Mongolian gerbils against ischemic injury: after-stroke-effect. Neurochem Res. 2005; 30: 1283–8.
  • Fujii T, Takaoka M, Tsuruoka N, Kiso Y, Tanaka T, Matsumura Y. Dietary supplementation of L-carnosine prevents ischemia/reperfusion-induced renal injury in rats. Biol Pharm Bull. 2005; 28: 361–3.
  • Kurata H, Fujii T, Tsutsui H, Katayama T, Ohkita M, Takaoka M, Tsuruoka N, Kiso Y, Ohno Y, Fujisawa Y, Shokoji T, Nishiyama A, Abe Y, Matsumura Y. Renoprotective effects of L-carnosine on ischemia/reperfusion-induced renal injury in rats. J Pharmacol Exp Ther. 2006; 319: 640–7.
  • Fouad AA, El-Rehany MA, Maghraby HK. The hepatoprotective effect of carnosine against ischemia/reperfusion liver injury in rats. Eur J Pharmacol. 2007; 572: 61–8.
  • Rajanikant GK, Zemke D, Senut MC, Frenkel MB, Chen AF, Gupta R, Majid A. Carnosine is neuroprotective against permanent focal cerebral ischemia in mice. Stroke. 2007; 38: 3023–31.
  • Baykara B, Tekmen I, Pekcetin C, Ulukus C, Tuncel P, Sagol O, Ormen M, Ozogul C. The protective effects of carnosine and melatonin in ischemia-reperfusion injury in the rat liver. Acta Histochem. 2009; 111: 42–51.
  • Pekcetin C, Kiray M, Ergur BU, Tugyan K, Bagriyanik HA, Erbil G, Baykara B, Camsari UM. Carnosine attenuates oxidative stress and apoptosis in transient cerebral ischemia in rats. Acta Biol Hung. 2009; 60: 137–48.
  • Sewerynek E, Reiter RJ, Melchiorri D, Ortiz GG, Lewinski A. Oxidative damage in the liver induced by ischemia-reperfusion: protection by melatonin. Hepatogastroenterology. 1996; 43: 898–905.
  • Cho S, Joh TH, Baik HH, Dibinis C, Volpe BT.1997; 755: 335–8.
  • De La Lastra CA, Cabeza J, Motilva V, Martin MJ. Melatonin protects against gastric ischemia-reperfusion injury in rats. J Pineal Res. 1997; 23: 47–52.
  • Guerrero JM, Reiter RJ, Ortiz GG, Pablos MI, Sewerynek E, Chuang JI. Melatonin prevents increases in neural nitric oxide and cyclic GMP production after transient brain ischemia and reperfusion in the Mongolian gerbil (Meriones unguiculatus). J Pineal Res. 1997; 23: 24–31.
  • Konturek PC, Konturek SJ, Majka J, Zembala M, Hahn EG. Melatonin affords protection against gastric lesions induced by ischemia-reperfusion possibly due to its antioxidant and mucosal microcirculatory effects. Eur J Pharmacol. 1997; 322: 73–7.
  • Li XJ, Zhang LM, Gu J, Zhang AZ, Sun FY. Melatonin decreases production of hydroxyl radical during cerebral ischemia-reperfusion. Zhongguo Yao Li Xue Bao. 1997; 18: 394–6.
  • Tan DX, Manchester LC, Reiter RJ, Qi W, Kim SJ, El-Sokkary GH. Ischemia/reperfusion-induced arrhythmias in the isolated rat heart: prevention by melatonin. J Pineal Res. 1998; 25: 184–91.
  • Kilic E, Ozdemir YG, Bolay H, Kele⋅timur H, Dalkara T. Pinealectomy aggravates and melatonin administration attenuates brain damage in focal ischemia. J Cereb Blood Flow Metab. 1999; 19: 511–6.
  • Ling X, Zhang LM, Lu SD, Li XJ, Sun FY. Protective effect of melatonin on injuried cerebral neurons is associated with bcl-2 protein over-expression. Zhongguo Yao Li Xue Bao. 1999; 20: 409–14.
  • Wakatsuki A, Okatani Y, Izumiya C, Ikenoue N. Melatonin protects against ischemia and reperfusion-induced oxidative lipid and DNA damage in fetal rat brain. J Pineal Res. 1999; 26: 147–52.
  • Borlongan CV, Yamamoto M, Takei N, Kumazaki M, Ungsuparkorn C, Hida H, Sanberg PR, Nishino H. Glial cell survival is enhanced during melatonin-induced neuroprotection against cerebral ischemia. FASEB J. 2000; 14: 1307–17.
  • Cuzzocrea S, Costantino G, Gitto E, Mazzon E, Fulia F, Serraino I, Cordaro S, Barberi I, De Sarro A, Caputi AP. Protective effects of melatonin in ischemic brain injury. J Pineal Res. 2000; 29: 217–27.
  • Cuzzocrea S, Costantino G, Mazzon E, Micali A, De Sarro A, Caputi AP. Beneficial effects of melatonin in a rat model of splanchnic artery occlusion and reperfusion. J Pineal Res. 2000; 28: 52–63.
  • Kaneko S, Okumura K, Numaguchi Y, Matsui H, Murase K, Mokuno S, Morishima I, Hira K, Toki Y, Ito T, Hayakawa T. Melatonin scavenges hydroxyl radical and protects isolated rat hearts from ischemic reperfusion injury. Life Sci. 2000; 67: 101–12.
  • Kazez A, Demirbağ M, Ustündağ B, Ozercan IH, Sağlam M. The role of melatonin in prevention of intestinal ischemia-reperfusion injury in rats. J Pediatr Surg. 2000; 35: 1444–8.
  • Lagneux C, Joyeux M, Demenge P, Ribuot C, Godin-Ribuot D. Protective effects of melatonin against ischemia-reperfusion injury in the isolated rat heart. Life Sci. 2000; 66: 503–9.
  • Cabeza J, Motilva V, Martín MJ, de la Lastra CA. Mechanisms involved in gastric protection of melatonin against oxidant stress by ischemia-reperfusion in rats. Life Sci. 2001; 68: 1405–15.
  • Okatani Y, Wakatsuki A, Shinohara K, Taniguchi K, Fukaya T. Melatonin protects against oxidative mitochondrial damage induced in rat placenta by ischemia and reperfusion. J Pineal Res. 2001; 31: 173–8.
  • Rodríguez-Reynoso S, Leal C, Portilla E, Olivares N, Muñiz J. Effect of exogenous melatonin on hepatic energetic status during ischemia/reperfusion: possible role of tumor necrosis factor-alpha and nitric oxide. J Surg Res. 2001; 100: 141–9.
  • Szárszoi O, Asemu G, Vanecek J, Ost’ádal B, Kolár F. Effects of melatonin on ischemia and reperfusion injury of the rat heart. Cardiovasc Drugs Ther. 2001; 15: 251–7.
  • Sinha K, Degaonkar MN, Jagannathan NR, Gupta YK. Effect of melatonin on ischemia reperfusion injury induced by middle cerebral artery occlusion in rats. Eur J Pharmacol. 2001; 428: 185–92.
  • Wakatsuki A, Okatani Y, Shinohara K, Ikenoue N, Fukaya T. Melatonin protects against ischemia/reperfusion-induced oxidative damage to mitochondria in fetal rat brain. J Pineal Res. 2001; 31: 167–72.
  • Celebi S, Dilsiz N, Yilmaz T, Kükner AS. Effects of melatonin, vitamin E and octreotide on lipid peroxidation during ischemia-reperfusion in the guinea pig retina. Eur J Ophthalmol. 2002; 12: 77–83.
  • El-Abhar HS, Shaalan M, Barakat M, El-Denshary ES. Effect of melatonin and nifedipine on some antioxidant enzymes and different energy fuels in the blood and brain of global ischemic rats. J Pineal Res. 2002; 33: 87–94.
  • Gupta YK, Chaudhary G, Sinha K. Enhanced protection by melatonin and meloxicam combination in a middle cerebral artery occlusion model of acute ischemic stroke in rat. Can J Physiol Pharmacol. 2002; 80: 210–7.
  • Kondoh T, Uneyama H, Nishino H, Torii K. Melatonin reduces cerebral edema formation caused by transient forebrain ischemia in rats. Life Sci. 2002; 72: 583–90.
  • Lee YM, Chen HR, Hsiao G, Sheu JR, Wang JJ, Yen MH. Protective effects of melatonin on myocardial ischemia/reperfusion injury in vivo. J Pineal Res. 2002; 33: 72–80.
  • Pei Z, Ho HT, Cheung RT. Pre-treatment with melatonin reduces volume of cerebral infarction in a permanent middle cerebral artery occlusion stroke model in the rat. Neurosci Lett. 2002; 318: 141–4.
  • Pei Z, Pang SF, Cheung RT. Pretreatment with melatonin reduces volume of cerebral infarction in a rat middle cerebral artery occlusion stroke model. J Pineal Res. 2002; 32: 168–72.
  • Reiter RJ, Tan DX, Sainz RM, Mayo JC. Melatonin protects the heart against both ischemia/reperfusion injury and chemotherapeutic drugs. Cardiovasc Drugs Ther. 2002; 16: 5–6.
  • Sahna E, Acet A, Ozer MK, Olmez E. Myocardial ischemia-reperfusion in rats: reduction of infarct size by either supplemental physiological or pharmacological doses of melatonin. J Pineal Res. 2002; 33: 234–8.
  • Sahna E, Olmez E, Acet A. Effects of physiological and pharmacological concentrations of melatonin on ischemia-reperfusion arrhythmias in rats: can the incidence of sudden cardiac death be reduced?. J Pineal Res. 2002; 32: 194–8.
  • Sener G, Paskaloğlu K, Sehirli AO, Dülger GA, Alican I. The effects of melatonin on ischemia-reperfusion induced changes in rat corpus cavernosum. J Urol. 2002; 167: 2624–7.
  • Sener G, Sehirli AO, Keyer-Uysal M, Arbak S, Ersoy Y, Yeğen BC. The protective effect of melatonin on renal ischemia-reperfusion injury in the rat. J Pineal Res. 2002; 32: 120–6.
  • Sun FY, Lin X, Mao LZ, Ge WH, Zhang LM, Huang YL, Gu J. Neuroprotection by melatonin against ischemic neuronal injury associated with modulation of DNA damage and repair in the rat following a transient cerebral ischemia. J Pineal Res. 2002; 33: 48–56.
  • Zhang J, Guo JD, Xing SH, Gu SL, Dai TJ. [The protective effects of melatonin on global cerebral ischemia-reperfusion injury in gerbils]. [Article in Chinese]. Yao Xue Xue Bao. 2002; 37: 329–33.
  • Cheung RT. The utility of melatonin in reducing cerebral damage resulting from ischemia and reperfusion. J Pineal Res. 2003; 34: 153–60.
  • Dobsak P, Siegelova J, Eicher JC, Jancik J, Svacinova H, Vasku J, Kuchtickova S, Horky M, Wolf JE.2003; 9: 179–187.
  • Erten SF, Kocak A, Ozdemir I, Aydemir S, Colak A, Reeder BS. Protective effect of melatonin on experimental spinal cord ischemia. Spinal Cord. 2003; 41: 533–8.
  • Kunduzova OR, Escourrou G, Seguelas MH, Delagrange P, De La Farge F, Cambon C, Parini A. Prevention of apoptotic and necrotic cell death, caspase-3 activation, and renal dysfunction by melatonin after ischemia/reperfusion. FASEB J. 2003; 17: 872–4.
  • Okatani Y, Wakatsuki A, Reiter RJ, Enzan H, Miyahara Y. Protective effect of melatonin against mitochondrial injury induced by ischemia and reperfusion of rat liver. Eur J Pharmacol. 2003; 469: 145–52.
  • Ozturk A, Baltaci AK, Mogulkoc R, Ozturk B. The effect of prophylactic melatonin administration on reperfusion damage in experimental testis ischemia-reperfusion. Neuro Endocrinol Lett. 2003; 24: 170–2.
  • Pei Z, Cheung RT. Melatonin protects SHSY5Y neuronal cells but not cultured astrocytes from ischemia due to oxygen and glucose deprivation. J Pineal Res. 2003; 34: 194–201.
  • Pei Z, Fung PC, Cheung RT. Melatonin reduces nitric oxide level during ischemia but not blood-brain barrier breakdown during reperfusion in a rat middle cerebral artery occlusion stroke model. J Pineal Res. 2003; 34: 110–8.
  • Pei Z, Pang SF, Cheung RT. Administration of melatonin after onset of ischemia reduces the volume of cerebral infarction in a rat middle cerebral artery occlusion stroke model. Stroke. 2003; 34: 770–5.
  • Reiter RJ, Sainz RM, Lopez-Burillo S, Mayo JC, Manchester LC, Tan DX. Melatonin ameliorates neurologic damage and neurophysiologic deficits in experimental models of stroke. Ann N Y Acad Sci. 2003;993:35–47; discussion48–53.
  • Reiter RJ, Tan DX. Melatonin: a novel protective agent against oxidative injury of the ischemic/reperfused heart. Cardiovasc Res. 2003; 58: 10–9.
  • Sahna E, Parlakpinar H, Ozturk F, Cigremis Y, Acet A. The protective effects of physiological and pharmacological concentrations of melatonin on renal ischemia-reperfusion injury in rats. Urol Res. 2003; 31: 188–93.
  • Sener G, Sehirli AO, Paskaloğlu K, Dülger GA, Alican I. Melatonin treatment protects against ischemia/reperfusion-induced functional and biochemical changes in rat urinary bladder. J Pineal Res. 2003; 34: 226–30.
  • Sener G, Tosun O, Sehirli AO, Kaçmaz A, Arbak S, Ersoy Y, Ayanoğlu-Dülger G. Melatonin and N-acetylcysteine have beneficial effects during hepatic ischemia and reperfusion. Life Sci. 2003; 72: 2707–18.
  • Abasiyanik A, Dağdönderen L. Beneficial effects of melatonin compared with allopurinol in experimental testicular torsion. J Pediatr Surg. 2004; 39: 1238–41.
  • Andrabi SA, Sayeed I, Siemen D, Wolf G, Horn TF. Direct inhibition of the mitochondrial permeability transition pore: a possible mechanism responsible for anti-apoptotic effects of melatonin. FASEB J. 2004; 18: 869–71.
  • Gupta S, Kaul CL, Sharma SS. Neuroprotective effect of combination of poly (ADP-ribose) polymerase inhibitor and antioxidant in middle cerebral artery occlusion induced focal ischemia in rats. Neurol Res. 2004; 26: 103–7.
  • Kilic E, Kilic U, Reiter RJ, Bassetti CL, Hermann DM. Prophylactic use of melatonin protects against focal cerebral ischemia in mice: role of endothelin converting enzyme-1. J Pineal Res. 2004; 37: 247–51.
  • Kilic E, Kilic U, Yulug B, Hermann DM, Reiter RJ. Melatonin reduces disseminate neuronal death after mild focal ischemia in mice via inhibition of caspase-3 and is suitable as an add-on treatment to tissue-plasminogen activator. J Pineal Res. 2004; 36: 171–6.
  • Lee EJ, Wu TS, Lee MY, Chen TY, Tsai YY, Chuang JI, Chang GL. Delayed treatment with melatonin enhances electrophysiological recovery following transient focal cerebral ischemia in rats. J Pineal Res. 2004; 36: 33–42.
  • Pei Z, Cheung RT. Pretreatment with melatonin exerts anti-inflammatory effects against ischemia/reperfusion injury in a rat middle cerebral artery occlusion stroke model. J Pineal Res. 2004; 37: 85–91.
  • Rodríguez-Reynoso S, Leal C, Portilla-de Buen E, Castillo JC, Ramos-Solano F. Melatonin ameliorates renal ischemia/reperfusion injury. J Surg Res. 2004; 116: 242–7.
  • Torii K, Uneyama H, Nishino H, Kondoh T. Melatonin suppresses cerebral edema caused by middle cerebral artery occlusion/reperfusion in rats assessed by magnetic resonance imaging. J Pineal Res. 2004; 36: 18–24.
  • Turkoz Y, Celik O, Hascalik S, Cigremis Y, Hascalik M, Mizrak B, Yologlu S. Melatonin reduces torsion-detorsion injury in rat ovary: biochemical and histopathologic evaluation. J Pineal Res. 2004; 37: 137–41.
  • Watanabe K, Wakatsuki A, Shinohara K, Ikenoue N, Yokota K, Fukaya T. Maternally administered melatonin protects against ischemia and reperfusion-induced oxidative mitochondrial damage in premature fetal rat brain. J Pineal Res. 2004; 37: 276–80.
  • Arslan SO, Gelir E, Sayan H, Ozacmak VH. L-Arginine and melatonin interaction in rat intestinal ischemia-reperfusion. Fundam Clin Pharmacol. 2005; 19: 533–5.
  • Erkanli K, Kayalar N, Erkanli G, Ercan F, Sener G, Kirali K. Melatonin protects against ischemia/reperfusion injury in skeletal muscle. J Pineal Res. 2005; 39: 238–42.
  • Eşrefoğlu M, Gül M, Parlakpinar H, Acet A. Effects of melatonin and caffeic acid phenethyl ester on testicular injury induced by myocardial ischemia/reperfusion in rats. Fundam Clin Pharmacol. 2005; 19: 365–72.
  • Kaçmaz A, User EY, Sehirli AO, Tilki M, Ozkan S, Sener G. Protective effect of melatonin against ischemia/reperfusion-induced oxidative remote organ injury in the rat. Surg Today. 2005; 35: 744–50.
  • Kilic E, Kilic U, Reiter RJ, Bassetti CL, Hermann DM. Tissue-plasminogen activator-induced ischemic brain injury is reversed by melatonin: role of iNOS and Akt. J Pineal Res. 2005; 39: 151–5.
  • Kilic U, Kilic E, Reiter RJ, Bassetti CL, Hermann DM. Signal transduction pathways involved in melatonin-induced neuroprotection after focal cerebral ischemia in mice. J Pineal Res. 2005; 38: 67–71.
  • Lee EJ, Lee MY, Chen HY, Hsu YS, Wu TS, Chen ST, Chang GL. Melatonin attenuates gray and white matter damage in a mouse model of transient focal cerebral ischemia. J Pineal Res. 2005; 38: 42–52.
  • Ozacmak VH, Sayan H, Arslan SO, Altaner S, Aktas RG.2005; 76: 1575–88.
  • Reiter RJ, Tan DX, Leon J, Kilic U, Kilic E. When melatonin gets on your nerves: its beneficial actions in experimental models of stroke. Exp Biol Med (Maywood). 2005; 230: 104–17.
  • Sahna E, Parlakpinar H, Turkoz Y, Acet A. Protective effects of melatonin on myocardial ischemia/reperfusion induced infarct size and oxidative changes. Physiol Res. 2005; 54: 491–5.
  • Tütüncüler F, Eskiocak S, Başaran UN, Ekuklu G, Ayvaz S, Vatansever U. The protective role of melatonin in experimental hypoxic brain damage. Pediatr Int. 2005; 47: 434–9.
  • Vazan R, Pancza D, Béder I, Styk J. Ischemia-reperfusion injury-antiarrhythmic effect of melatonin associated with reduced recovering of contractility. Gen Physiol Biophys. 2005; 24: 355–9.
  • Chen HY, Chen TY, Lee MY, Chen ST, Hsu YS, Kuo YL, Chang GL, Wu TS, Lee EJ. Melatonin decreases neurovascular oxidative/nitrosative damage and protects against early increases in the blood-brain barrier permeability after transient focal cerebral ischemia in mice. J Pineal Res. 2006; 41: 175–82.
  • Chen TY, Lee MY, Chen HY, Kuo YL, Lin SC, Wu TS, Lee EJ. Melatonin attenuates the postischemic increase in blood-brain barrier permeability and decreases hemorrhagic transformation of tissue-plasminogen activator therapy following ischemic stroke in mice. J Pineal Res. 2006; 40: 242–50.
  • Duan Q, Wang Z, Lu T, Chen J, Wang X. Comparison of 6-hydroxylmelatonin or melatonin in protecting neurons against ischemia/reperfusion-mediated injury. J Pineal Res. 2006; 41: 351–7.
  • Gurlek A, Celik M, Parlakpinar H, Aydogan H, Bay-Karabulut A. The protective effect of melatonin on ischemia-reperfusion injury in the groin (inferior epigastric) flap model in rats. J Pineal Res. 2006; 40: 312–7.
  • Han YX, Zhang SH, Wang XM, Wu JB. Inhibition of mitochondria responsible for the anti-apoptotic effects of melatonin during ischemia-reperfusion. J Zhejiang Univ Sci B. 2006; 7: 142–7.
  • Lochner A, Genade S, Davids A, Ytrehus K, Moolman JA. Short- and long-term effects of melatonin on myocardial post-ischemic recovery. J Pineal Res. 2006; 40: 56–63.
  • Muñoz-Casares FC, Padillo FJ, Briceño J, Collado JA, Muñoz-Castañeda JR, Ortega R, Cruz A, Túnez I, Montilla P, Pera C, Muntané J. Melatonin reduces apoptosis and necrosis induced by ischemia/reperfusion injury of the pancreas. J Pineal Res. 2006; 40: 195–203.
  • Petrosillo G, Di Venosa N, Pistolese M, Casanova G, Tiravanti E, Colantuono G, Federici A, Paradies G, Ruggiero FM. Protective effect of melatonin against mitochondrial dysfunction associated with cardiac ischemia- reperfusion: role of cardiolipin. FASEB J. 2006; 20: 269–76.
  • Sahach VF, Rudyk OV, Vavilova HL, Kotsiuruba AV, Tkachenko IuP. [Melatonin recovers ischemic tolerance and decreases the sensitivity of mitochondrial permeability transition pore opening in the heart of aging rats]. [Article in Ukrainian]. Fiziol Zh. 2006;52: 3–14.
  • Sener G, Sert G, Ozer Sehirli A, Arbak S, Gedik N, Ayanoğlu-Dülger G. Melatonin protects against pressure ulcer-induced oxidative injury of the skin and remote organs in rats. J Pineal Res. 2006; 40: 280–7.
  • Zhang WH, Li JY, Zhou Y. Melatonin abates liver ischemia/reperfusion injury by improving the balance between nitric oxide and endothelin. Hepatobiliary Pancreat Dis Int. 2006; 5: 574–9.
  • Zou LY, Cheung RT, Liu S, Li G, Huang L. Melatonin reduces infarction volume in a photothrombotic stroke model in the wild-type but not cyclooxygenase-1-gene knockout mice. J Pineal Res. 2006; 41: 150–6.
  • Aktoz T, Aydogdu N, Alagol B, Yalcin O, Huseyinova G, Atakan IH. The protective effects of melatonin and vitamin E against renal ischemia-reperfusion injury in rats. Ren Fail. 2007; 29: 535–42.
  • Colak C, Parlakpinar H, Ozer MK, Sahna E, Cigremis Y, Acet A. Investigating the protective effect of melatonin on liver injury related to myocardial ischemia-reperfusion. Med Sci Monit. 2007; 13: BR251–254.
  • Kurcer Z, Oguz E, Ozbilge H, Baba F, Aksoy N, Celik H, Cakir H, Gezen MR. Melatonin protects from ischemia/reperfusion-induced renal injury in rats: this effect is not mediated by proinflammatory cytokines. J Pineal Res. 2007; 43: 172–8.
  • Lee MY, Kuan YH, Chen HY, Chen TY, Chen ST, Huang CC, Yang IP, Hsu YS, Wu TS, Lee EJ. Intravenous administration of melatonin reduces the intracerebral cellular inflammatory response following transient focal cerebral ischemia in rats. J Pineal Res. 2007; 42: 297–309.
  • Park SW, Choi SM, Lee SM. Effect of melatonin on altered expression of vasoregulatory genes during hepatic ischemia/reperfusion. Arch Pharm Res. 2007; 30: 1619–24.
  • Reiter RJ, Tan DX, Manchester LC, Tamura H. Melatonin defeats neurally-derived free radicals and reduces the associated neuromorphological and neurobehavioral damage. J Physiol Pharmacol. 2007; 58(Suppl 6): 5–22.
  • Sahna E, Türk G, Atessahin A, Yilmaz S, Olmez E. Remote organ injury induced by myocardial ischemia and reperfusion on reproductive organs, and protective effect of melatonin in male rats. Fertil Steril. 2007; 88: 188–92.
  • Welin AK, Svedin P, Lapatto R, Sultan B, Hagberg H, Gressens P, Kjellmer I, Mallard C. Melatonin reduces inflammation and cell death in white matter in the mid-gestation fetal sheep following umbilical cord occlusion. Pediatr Res. 2007; 61: 153–8.
  • Cervantes M, Moralí G, Letechipía-Vallejo G. Melatonin and ischemia-reperfusion injury of the brain. J Pineal Res. 2008; 45: 1–7.
  • Ceyran H, Narin F, Narin N, Akgun H, Ceyran AB, Ozturk F, Akcali Y. The effect of high dose melatonin on cardiac ischemia- reperfusion Injury. Yonsei Med J. 2008; 49: 735–41.
  • Fadillioglu E, Kurcer Z, Parlakpinar H, Iraz M, Gursul C. Melatonin treatment against remote organ injury induced by renal ischemia reperfusion injury in diabetes mellitus. Arch Pharm Res. 2008; 31: 705–12.
  • Genade S, Genis A, Ytrehus K, Huisamen B, Lochner A. Melatonin receptor-mediated protection against myocardial ischaemia/reperfusion injury: role of its anti-adrenergic actions. J Pineal Res. 2008; 45: 449–58.
  • Hung YC, Chen TY, Lee EJ, Chen WL, Huang SY, Lee WT, Lee MY, Chen HY, Wu TS. Melatonin decreases matrix metalloproteinase-9 activation and expression and attenuates reperfusion-induced hemorrhage following transient focal cerebral ischemia in rats. J Pineal Res. 2008; 45: 459–67.
  • Jiang CL, Yang BX, Zhao D, Jia R. [Total hepatic ischemia-reperfusion-induced lung injury in rats and protective effects of melatonin]. [Article in Chinese]. Beijing Da Xue Xue Bao. 2008; 40: 285–91.
  • Kim SH, Lee SM. Cytoprotective effects of melatonin against necrosis and apoptosis induced by ischemia/reperfusion injury in rat liver. J Pineal Res. 2008; 44: 165–71.
  • Koh PO. Melatonin regulates nitric oxide synthase expression in ischemic brain injury. J Vet Med Sci. 2008; 70: 747–50.
  • Korkmaz A, Oyar EO, Kardeş O, Omeroğlu S.2008; 5: 46–51.
  • Kurcer Z, Oguz E, Ozbilge H, Baba F, Aksoy N, Celik N. Effect of melatonin on testicular ischemia/reperfusion injury in rats: is this effect related to the proinflammatory cytokines?. Fertil Steril. 2008; 89((5 Suppl)): 1468–73.
  • Li JY, Yin HZ, Gu X, Zhou Y, Zhang WH, Qin YM. Melatonin protects liver from intestine ischemia reperfusion injury in rats. World J Gastroenterol. 2008; 14: 7392–6.
  • Li ZR, Shen MH, Niu WM. [Involvement of melatonin in the adjusting effect of electroacupuncture in resisting oxygen stress in cerebral ischemia-reperfusion injury rats]. [Article in Chinese]. Zhen Ci Yan Jiu. 2008; 33: 164–8.
  • Nagai R, Watanabe K, Wakatsuki A, Hamada F, Shinohara K, Hayashi Y, Imamura R, Fukaya T. Melatonin preserves fetal growth in rats by protecting against ischemia/reperfusion-induced oxidative/nitrosative mitochondrial damage in the placenta. J Pineal Res. 2008; 45: 271–6.
  • Sahna E, Deniz E, Bay-Karabulut A, Burma O. Melatonin protects myocardium from ischemia-reperfusion injury in hypertensive rats: role of myeloperoxidase activity. Clin Exp Hypertens. 2008; 30: 673–81.
  • Yeung HM, Hung MW, Fung ML. Melatonin ameliorates calcium homeostasis in myocardial and ischemia-reperfusion injury in chronically hypoxic rats. J Pineal Res. 2008; 45: 373–82.
  • Yurtçu M, Abasiyanik A, Avunduk MC, Muhtaroğlu S. Effects of melatonin on spermatogenesis and testicular ischemia-reperfusion injury after unilateral testicular torsion-detorsion. J Pediatr Surg. 2008; 43: 1873–8.
  • Chen HY, Hung YC, Chen TY, Huang SY, Wang YH, Lee WT, Wu TS, Lee EJ. Melatonin improves presynaptic protein, SNAP-25, expression and dendritic spine density and enhances functional and electrophysiological recovery following transient focal cerebral ischemia in rats. J Pineal Res. 2009; 47: 260–70.
  • Chen Z, Chua CC, Gao J, Chua KW, Ho YS, Hamdy RC, Chua BH. Prevention of ischemia/reperfusion-induced cardiac apoptosis and injury by melatonin is independent of glutathione peroxidase 1. J Pineal Res. 2009; 46: 235–41.
  • Diez ER, Prados LV, Carrión A, Ponce ZA, Miatello RM. A novel electrophysiologic effect of melatonin on ischemia/reperfusion-induced arrhythmias in isolated rat hearts. J Pineal Res. 2009; 46: 155–60.
  • Dominguez-Rodriguez A, Abreu-Gonzalez P, Reiter RJ. Clinical aspects of melatonin in the acute coronary syndrome. Curr Vasc Pharmacol. 2009; 7: 367–73.
  • Kesik V, Guven A, Vurucu S, Tunc T, Uysal B, Gundogdu G, Oztas E, Korkmaz A. Melatonin and 1400 W ameliorate both intestinal and remote organ injury following mesenteric ischemia/reperfusion. J Surg Res. 2009; 157: e97–e105.
  • Li Z, Nickkholgh A, Yi X, Bruns H, Gross ML, Hoffmann K, Mohr E, Zorn M, Büchler MW, Schemmer P. Melatonin protects kidney grafts from ischemia/reperfusion injury through inhibition of NF-kB and apoptosis after experimental kidney transplantation. J Pineal Res. 2009; 46: 365–72.
  • Liang R, Nickkholgh A, Hoffmann K, Kern M, Schneider H, Sobirey M, Zorn M, Büchler MW, Schemmer P. Melatonin protects from hepatic reperfusion injury through inhibition of IKK and JNK pathways and modification of cell proliferation. J Pineal Res. 2009; 46: 8–14.
  • Ozbek E, Ilbey YO, Ozbek M, Simsek A, Cekmen M, Somay A. Melatonin attenuates unilateral ureteral obstruction-induced renal injury by reducing oxidative stress, iNOS, MAPK, and NF-kB expression. J Endourol. 2009; 23: 1165–73.
  • Ozacmak VH, Barut F, Ozacmak HS. Melatonin provides neuroprotection by reducing oxidative stress and HSP70 expression during chronic cerebral hypoperfusion in ovariectomized rats. J Pineal Res. 2009; 47: 156–63.
  • Petrosillo G, Colantuono G, Moro N, Ruggiero FM, Tiravanti E, Di Venosa N, Fiore T, Paradies G. Melatonin protects against heart ischemia-reperfusion injury by inhibiting mitochondrial permeability transition pore opening. Am J Physiol Heart Circ Physiol. 2009; 297: H1487–93.
  • Wang X, Figueroa BE, Stavrovskaya IG, Zhang Y, Sirianni AC, Zhu S, Day AL, Kristal BS, Friedlander RM. Methazolamide and melatonin inhibit mitochondrial cytochrome c release and are neuroprotective in experimental models of ischemic injury. Stroke. 2009; 40: 1877–85.
  • Esposito E, Cuzzocrea S. Antiinflammatory activity of melatonin in central nervous system. Curr Neuropharmacol. 2010; 8: 228–42.
  • Dominguez-Rodriguez A, Abreu-Gonzalez P. Myocardial ischemia-reperfusion injury: Possible role of melatonin. World J Cardiol. 2010; 2: 233–6.
  • Hamada F, Watanabe K, Wakatsuki A, Nagai R, Shinohara K, Hayashi Y, Imamura R, Fukaya T. Therapeutic effects of maternal melatonin administration on ischemia/reperfusion-induced oxidative cerebral damage in neonatal rats. Neonatology. 2010; 98: 33–40.
  • Park OK, Yoo KY, Lee CH, Choi JH, Hwang IK, Park JH, Kwon YG, Kim YM, Won MH. Arylalkylamine N-acetyltransferase (AANAT) is expressed in astrocytes and melatonin treatment maintains AANAT in the gerbil hippocampus induced by transient cerebral ischemia. J Neurol Sci. 2010; 294: 7–17.
  • Reiter RJ, Tan DX, Paredes SD, Fuentes-Broto L. Beneficial effects of melatonin in cardiovascular disease. Ann Med. 2010; 42: 276–85.
  • Tai SH, Chen HY, Lee EJ, Chen TY, Lin HW, Hung YC, Huang SY, Chen YH, Lee WT, Wu TS. Melatonin inhibits postischemic matrix metalloproteinase-9 (MMP-9) activation via dual modulation of plasminogen/plasmin system and endogenous MMP inhibitor in mice subjected to transient focal cerebral ischemia. J Pineal Res. 2010; 49: 332–41.
  • Du To itEF F, Blackhurst D, Marais D, Lochner A. Chronic melatonin consumption prevents obesity-related metabolic abnormalities and protects the heart against myocardial ischemia and reperfusion injury in a prediabetic model of diet-induced obesity. J Pineal Res. 2011; 50: 171–82.
  • Brune D, Hellborg R, Persson BRR, Pääkkönen R. Radiation at Home, Outdoors and in the Workplace. Scandinavian Science Publishers. Oslo 2001.
  • Aruoma OI, Halliwell B, Hoey BM, Butler J. The antioxidant action of taurine, hypotaurine and their metabolic precursors. Biochem J. 1988; 256: 251–5.
  • Mehta TR, Dawson R Jr. Taurine is a weak scavenger of peroxynitrite and does not attenuate sodium nitroprusside toxicity to cells in culture. Amino Acids. 2001; 20: 419–33.
  • Oliveira MW, Minotto JB, de Oliveira MR, Zanotto-Filho A, Behr GA, Rocha RF, Moreira JC, Klamt F. Scavenging and antioxidant potential of physiological taurine concentrations against different reactive oxygen/nitrogen species. Pharmacol Rep. 2010; 62: 185–93.
  • Pryor WA. The role of free radical reactions in biological systems. In: Pryor WA. Free Radicals in Biology. Vol 1. Academic Press., New York, 1976, pp. 1–43.
  • Aust SD, Morehouse LA, Thomas CE. Role of metals in oxygen radical reactions. J Free Radic Biol Med. 1985; 1: 3–25.
  • Halliwell B, Gutteridge JM, Blake D. Metal ions and oxygen radical reactions in human inflammatory joint disease. Philos Trans R Soc Lond B Biol Sci. 1985; 311: 659–71.
  • Biemond P, Swaak AJ, Beindorff CM, Koster JF. Superoxide-dependent and –independent mechanisms of iron mobilization from ferritin by xanthine oxidase. Implications for oxygen-free-radical-induced tissue destruction during ischaemia and inflammation. Biochem J. 1986; 239: 169–73.
  • Biemond P, Swaak AJ, van Eijk HG, Koster JF. Intraarticular ferritin-bound iron in rheumatoid arthritis. A factor that increases oxygen free radical-induced tissue destruction. Arthritis Rheum. 1986; 29: 1187–93.
  • Biemond P, Swaak AJ, van Eijk HG, Koster JF. Superoxide dependent iron release from ferritin in inflammatory diseases. Free Radic Biol Med. 1988; 4: 185–98.
  • Patt A, Horesh IR, Berger EM, Harken AH, Repine JE. Iron depletion or chelation reduces ischemia/reperfusion-induced edema in gerbil brains. J Pediatr Surg. 1990;25:224–7; discussion227–8.
  • Garner B, Roberg K, Brunk UT. Endogenous ferritin protects cells with iron-laden lysosomes against oxidative stress. Free Radic Res. 1998; 29: 103–14.
  • Persson HL, Yu Z, Tirosh O, Eaton JW, Brunk UT. Prevention of oxidant-induced cell death by lysosomotropic iron chelators. Free Radic Biol Med. 2003; 34: 1295–305.
  • Yu Z, Eaton JW, Persson HL. The radioprotective agent, amifostine, suppresses the reactivity of intralysosomal iron. Redox Rep. 2003; 8: 347–55.
  • Yu Z, Persson HL, Eaton JW, Brunk UT. Intralysosomal iron: a major determinant of oxidant-induced cell death. Free Radic Biol Med. 2003; 34: 1243–52.
  • Valko M, Morris H, Cronin MT. Metals, toxicity and oxidative stress. Curr Med Chem. 2005; 12: 1161–208.
  • Kurz T, Gustafsson B, Brunk UT. Intralysosomal iron chelation protects against oxidative stress-induced cellular damage. FEBS J. 2006; 273: 3106–17.
  • Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem Biol Interact. 2006; 160: 1–40.
  • Jomova K, Valko M. Advances in metal-induced oxidative stress and human disease. Toxicology. 2011 Mar 14. [Epub ahead of print].
  • Claycamp HG, Luo D. Plutonium-catalyzed oxidative DNA damage in the absence of significant alpha-particle decay. Radiat Res. 1994; 137: 114–7.
  • Rodriguez H, Holmquist GP, D'Agostino R Jr, Keller J, Akman SA. Metal ion-dependent hydrogen peroxide-induced DNA damage is more sequence specific than metal specific. Cancer Res. 1997;57: 2394–403.
  • Kasprzak KS. Oxidative DNA and protein damage in metal-induced toxicity and carcinogenesis. Free Radic Biol Med. 2002; 32: 958–67.
  • Kawanishi S, Hiraku Y, Murata M, Oikawa S. The role of metals in site-specific DNA damage with reference to carcinogenesis. Free Radic Biol Med. 2002; 32: 822–32.
  • Lee DH, O'Connor TR, Pfeifer GP. Oxidative DNA damage induced by copper and hydrogen peroxide promotes CG- > TT tandem mutations at methylated CpG dinucleotides in nucleotide excision repair-deficient cells. Nucleic Acids Res. 2002; 30: 3566–73.
  • Miller AC, Stewart M, Brooks K, Shi L, Page N. Depleted uranium-catalyzed oxidative DNA damage: absence of significant alpha particle decay. J Inorg Biochem. 2002; 91: 246–52.
  • Kurz T, Leake A, Von Zglinicki T, Brunk UT. Relocalized redox-active lysosomal iron is an important mediator of oxidative-stress-induced DNA damage. Biochem J. 2004; 378: 1039–45.
  • Melidou M, Riganakos K, Galaris D. Protection against nuclear DNA damage offered by flavonoids in cells exposed to hydrogen peroxide: the role of iron chelation. Free Radic Biol Med. 2005; 39: 1591–600.
  • Battin EE, Perron NR, Brumaghim JL. The central role of metal coordination in selenium antioxidant activity. Inorg Chem. 2006; 45: 499–50.
  • Hong H, Cao H, Wang Y, Wang Y. Identification and quantification of a guanine-thymine intrastrand cross-link lesion induced by Cu(II)/H2O2/ascorbate. Chem Res Toxicol. 2006; 19: 614–21.
  • Cao H, Wang Y. Quantification of oxidative single-base and intrastrand cross-link lesions in unmethylated and CpG-methylated DNA induced by Fenton-type reagents. Nucleic Acids Res. 2007; 35: 4833–44.
  • Battin EE, Brumaghim JL. Metal specificity in DNA damage prevention by sulfur antioxidants. J Inorg Biochem. 2008; 102: 2036–42.
  • Cai C, Ching A, Lagace C, Linsenmayer T. Nuclear ferritin-mediated protection of corneal epithelial cells from oxidative damage to DNA. Dev Dyn. 2008; 237: 2676–83.
  • Perron NR, Hodges JN, Jenkins M, Brumaghim JL. Predicting how polyphenol antioxidants prevent DNA damage by binding to iron. Inorg Chem. 2008; 47: 6153–61.
  • Liu Y, Hu N. Electrochemical detection of natural DNA damage induced by ferritin/ascorbic acid/H2O2 system and amplification of DNA damage by endonuclease Fpg. Biosens Bioelectron. 2009; 25: 185–90.
  • Battin EE, Zimmerman MT, Ramoutar RR, Quarles CE, Brumaghim JL. Preventing metal-mediated oxidative DNA damage with selenium compounds. Metallomics. 2011. Feb 2. [Epub ahead of print].
  • Ayene IS, Koch CJ, Krisch RE. DNA strand breakage by bivalent metal ions and ionizing radiation. Int J Radiat Biol. 2007; 83: 195–210.
  • Berndt C, Kurz T, Selenius M, Fernandes AP, Edgren MR, Brunk UT. Chelation of lysosomal iron protects against ionizing radiation. Biochem J. 2010; 432: 295–301.
  • Svingen BA, Buege JA, O'Neal FO, Aust SD. The mechanism of NADPH-dependent lipid peroxidation. The propagation of lipid peroxidation. J Biol Chem. 1979; 254: 5892–9.
  • Aust SD, Svingen BA. The role of iron in enzymatic lipid peroxidation. In: Pryor WA. Free Radicals in Biology. Vol V. Academic Press., New York, 1982, pp. 1–28.
  • Sugioka K, Nakano M. Mechanism of phospholipid peroxidation induced by ferric ion-ADP-adriamycin-co-ordination complex. Biochim Biophys Acta. 1982; 713: 333–43.
  • Sugioka K, Nakano H, Nakano M, Tero-Kubota S, Ikegami Y. Generation of hydroxyl radicals during the enzymatic reductions of the Fe3 + -ADP-phosphate-adriamycin and Fe3 + -ADP-EDTA systems. Less involvement of hydroxyl radical and a great importance of proposed perferryl ion complexes in lipid peroxidation. Biochim Biophys Acta. 1983; 753: 411–21.
  • Nakano H, Sugioka K, Nakano M, Mizukami M, Kimura H, Tero-Kubota S, Ikegami Y. Importance of Fe2 + -ADP and the relative unimportance of OH in the mechanism of mitomycin C-induced lipid peroxidation. Biochim Biophys Acta. 1984; 796: 285–93.
  • Morehouse LA, Aust SD. Reconstituted microsomal lipid peroxidation: ADP-Fe3 + -dependent peroxidation of phospholipid vesicles containing NADPH-cytochrome P450 reductase and cytochrome P450. Free Radic Biol Med. 1988; 4: 269–77.
  • Goldschmidt VM. Geochemistry. Clarendon Press: Oxford, 1954
  • Gutteridge JM, Nagy I, Maidt L, Floyd RA. ADP-iron as a Fenton reactant: radical reactions detected by spin trapping, hydrogen abstraction, and aromatic hydroxylation. Arch Biochem Biophys. 1990; 277: 422–8.
  • Christophersen OA, Haug A. Possible roles of oxidative stress, local circulatory failure and nutrition factors in the patogenesis of hypervirulent influenza: implications for therapy and global emergency preparedness. Microbial Ecology in Health and Disease. 2005; 17: 189–99.
  • Hansen SH, Andersen ML, Birkedal H, Cornett C, Wibrand F. The important role of taurine in oxidative metabolism. Adv Exp Med Biol. 2006; 583: 129–35.
  • Hansen SH, Andersen ML, Cornett C, Gradinaru R, Grunnet N. A role for taurine in mitochondrial function. J Biomed Sci. 2010; 1(17 Suppl): S23.
  • Haug A, Rødbotten R, Mydland LT, Christophersen OA. Increased broiler muscle carnosine and anserine following histidine supplementation of commercial broiler feed concentrate. Acta Agriculturae Scandinavica. Section A – Animal Sciences. 2008; 58(2): 71–7.
  • Messina SA, Dawson R Jr. Attenuation of oxidative damage to DNA by taurine and taurine analogs. Adv Exp Med Biol. 2000; 483: 355–67.
  • Dawson R Jr, Baker D, Eppler B, Tang E, Shih D, Hern H, Hu M. Taurine inhibition of metal-stimulated catecholamine oxidation. Neurotox Res. 2000; 2: 1–15.
  • Biasetti M, Dawson R Jr. Effects of sulfur containing amino acids on iron and nitric oxide stimulated catecholamine oxidation. Amino Acids. 2002; 22: 351–68.
  • Rose RC, Bode AM. Biology of free radical scavengers: an evaluation of ascorbate. FASEB J. 1993; 7: 1135–42.
  • Casalino E, Sblano C, Landriscina C. A possible mechanism for initiation of lipid peroxidation by ascorbate in rat liver microsomes. Int J Biochem Cell Biol. 1996; 28: 137–49.
  • Bachowski GJ, Thomas JP, Girotti AW. Ascorbate-enhanced lipid peroxidation in photooxidized cell membranes: cholesterol product analysis as a probe of reaction mechanism. Lipids. 1988; 23: 580–6.
  • Laudicina DC, Marnett LJ. Enhancement of hydroperoxide-dependent lipid peroxidation in rat liver microsomes by ascorbic acid. Arch Biochem Biophys. 1990; 278: 73–80.
  • Miller DM, Aust SD. Studies of ascorbate-dependent, iron-catalyzed lipid peroxidation. Arch Biochem Biophys. 1989; 271: 113–9.
  • Christophersen OA, Haug A. More about hypervirulent avian influenza: is the world now better prepared?. Microb Ecol Health Dis. 2006; 19: 78–121.
  • Moxnes JF, Christophersen OA. The Spanish Flu as a worst case scenario?. Microb Ecol Health Dis. 2008; 20: 1–26.
  • Christophersen OA, Haug A. Animal products, diseases and drugs: a plea for better integration between agricultural sciences, human nutrition and human pharmacology. Lipids Health Dis. 2011 Jan 20;10:16.
  • Huxtable RJ. Physiological actions of taurine. Physiol Rev. 1992; 72: 101–63.
  • Seabra V, Stachlewitz RF, Thurman RG. Taurine blunts LPS-induced increases in intracellular calcium and TNF-alpha production by Kupffer cells. J Leukoc Biol. 1998; 64: 615–21.
  • Wheeler MD, Thurman RG. Production of superoxide and TNF-alpha from alveolar macrophages is blunted by glycine. Am J Physiol. 1999; 277: L952–9.
  • Wheeler MD, Ikejema K, Enomoto N, Stacklewitz RF, Seabra V, Zhong Z, Yin M, Schemmer P, Rose ML, Rusyn I, Bradford B, Thurman RG. Glycine: a new anti-inflammatory immunonutrient. Cell Mol Life Sci. 1999; 56: 843–56.
  • Kim SK, Kim YC. Attenuation of bacterial lipopolysaccharide-induced hepatotoxicity by betaine or taurine in rats. Food Chem Toxicol. 2002; 40: 545–9.
  • Wheeler MD, Stachlewitz RF, Yamashina S, Ikejima K, Morrow AL, Thurman RG. Glycine-gated chloride channels in neutrophils attenuate calcium influx and superoxide production. FASEB J. 2000; 14: 476–84.
  • Yanni G, Whelan A, Feighery C, Bresnihan B. Synovial tissue macrophages and joint erosion in rheumatoid arthritis. Ann Rheum Dis. 1994; 53: 39–44.
  • Burmester GR, Stuhlmüller B, Keyszer G, Kinne RW. Mononuclear phagocytes and rheumatoid synovitis. Mastermind or workhorse in arthritis?. Arthritis Rheum. 1997; 40: 5–18.
  • Bauerová K, Bezek A. Role of reactive oxygen and nitrogen species in etiopathogenesis of rheumatoid arthritis. Gen Physiol Biophys. 1999; 18: 15–20.
  • Bresnihan B. Pathogenesis of joint damage in rheumatoid arthritis. J Rheumatol. 1999; 26: 717–9.
  • Cunnane G, FitzGerald O, Hummel KM, Youssef PP, Gay RE, Gay S, Bresnihan B. Synovial tissue protease gene expression and joint erosions in early rheumatoid arthritis. Arthritis Rheum. 2001; 44: 1744–53.
  • Smeets TJ, Kraan MC, Galjaard S, Youssef PP, Smith MD, Tak PP. Analysis of the cell infiltrate and expression of matrix metalloproteinases and granzyme B in paired synovial biopsy specimens from the cartilage-pannus junction in patients with RA. Ann Rheum Dis. 2001; 60: 561–5.
  • Kraan MC, Reece RJ, Smeets TJ, Veale DJ, Emery P, Tak PP. Comparison of synovial tissues from the knee joints and the small joints of rheumatoid arthritis patients: Implications for pathogenesis and evaluation of treatment. Arthritis Rheum. 2002; 46: 2034–8.
  • Smeets TJ, Barg EC, Kraan MC, Smith MD, Breedveld FC, Tak PP. Analysis of the cell infiltrate and expression of proinflammatory cytokines and matrix metalloproteinases in arthroscopic synovial biopsies: comparison with synovial samples from patients with end stage, destructive rheumatoid arthritis. Ann Rheum Dis. 2003; 62: 635–8.
  • den Broeder AA, Wanten GJ, Oyen WJ, Naber T, van Riel PL, Barrera P. Neutrophil migration and production of reactive oxygen species during treatment with a fully human anti-tumor necrosis factor-alpha monoclonal antibody in patients with rheumatoid arthritis. J Rheumatol. 2003; 30: 232–7.
  • Opstvedt J, Olsen S, Urdahl N, Laksesvela B, Bjørnstad J. Næringsverdien av Fiskemel Produsert fra Ulike Fiskearter. [The Nutritive Value value of Fish Meal Produced from Different Species of Fish] [Report in Norwegian]. Meld. SSF (Sildolje- og Sildemelindustriens Forskningsinstitutt [Research Institute of the Norwegian Herring Oil and Meal Industry]. 1970; 4: 118–168.
  • Haug A, Christophersen OA, Kinabo J, Kaunda W, Eik LO. Use of dried kapenta and other products based on whole fish for complementing maize-based diets. AJFAND (Afr J Food Agric Nutr Dev). 2010; 10: 2478–2500.
  • Hajizadeh S, DeGroot J, TeKoppele JM, Tarkowski A, Collins LV. Extracellular mitochondrial DNA and oxidatively damaged DNA in synovial fluid of patients with rheumatoid arthritis. Arthritis Res Ther. 2003; 5: R234–40.
  • Yousefi S, Mihalache C, Kozlowski E, Schmid I, Simon HU. Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ. 2009; 16: 1438–44.
  • Hajizadeh S, DeGroot J, TeKoppele JM, Tarkowski A, Collins LV. Endogenously oxidized mitochondrial DNA induces in vivo and in vitro inflammatory responses. Arthritis Res Ther. 2003; 5: R234–40.
  • Zhang Q, Itagaki K, Hauser CJ. Mitochondrial DNA is released by shock and activates neutrophils via p38 MAP kinase. Shock. 2010; 34: 55–9.
  • Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, Brohi K, Itagaki K, Hauser CJ. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010; 464: 104–7.
  • Rabiet MJ, Huet E, Boulay F. Human mitochondria-derived N-formylated peptides are novel agonists equally active on FPR and FPRL1, while Listeria monocytogenes-derived peptides preferentially activate FPR. Eur J Immunol. 2005; 35: 2486–95.
  • Raoof M, Zhang Q, Itagaki K, Hauser CJ. Mitochondrial peptides are potent immune activators that activate human neutrophils via FPR-1. J Trauma. 2010; 68: 1328–32; discussion 1332-4.
  • Pullerits R, Bokarewa M, Jonsson IM, Verdrengh M, Tarkowski A. Extracellular cytochrome c, a mitochondrial apoptosis-related protein, induces arthritis. Rheumatology (Oxford). 2005; 44: 32–9.
  • Pullerits R, Jonsson IM, Verdrengh M, Bokarewa M, Andersson U, Erlandsson-Harris H, Tarkowski A. High mobility group box chromosomal protein 1, a DNA binding cytokine, induces arthritis. Arthritis Rheum. 2003; 48: 1693–700.
  • Fan J, Li Y, Levy RM, Fan JJ, Hackam DJ, Vodovotz Y, Yang H, Tracey KJ, Billiar TR, Wilson MA. Hemorrhagic shock induces NAD(P)H oxidase activation in neutrophils: role of HMGB1-TLR4 signaling. J Immunol. 2007; 178: 6573–80.
  • Tsung A, Klune JR, Zhang X, Jeyabalan G, Cao Z, Peng X, Stolz DB, Geller DA, Rosengart MR, Billiar TR. HMGB1 release induced by liver ischemia involves Toll-like receptor 4 dependent reactive oxygen species production and calcium-mediated signaling. J Exp Med. 2007; 204: 2913–23.
  • Pullerits R, Jonsson IM, Kollias G, Tarkowski A. Induction of arthritis by high mobility group box chromosomal protein 1 is independent of tumour necrosis factor signalling. Arthritis Res Ther. 2008; 10: R72.
  • Tang D, Kang R, Zeh HJ, Lotze MT. High-mobility group box 1, oxidative stress, and disease. Antioxid Redox Signal. 2011; 14: 1315–35.
  • Cillero-Pastor B, Martin MA, Arenas J, López-Armada MJ, Blanco FJ. Effect of nitric oxide on mitochondrial activity of human synovial cells. BMC Musculoskelet Disord. 2011; 12: 42.
  • López-Armada MJ, Caramés B, Martín MA, Cillero-Pastor B, Lires-Dean M, Fuentes-Boquete I, Arenas J, Blanco FJ. Mitochondrial activity is modulated by TNFalpha and IL-1beta in normal human chondrocyte cells. Osteoarthritis Cartilage. 2006; 14: 1011–22.
  • Mariappan N, Elks CM, Fink B, Francis J. TNF-induced mitochondrial damage: a link between mitochondrial complex I activity and left ventricular dysfunction. Free Radic Biol Med. 2009; 46: 462–70.
  • Kim J, Xu M, Xo R, Mates A, Wilson GL, Pearsall AW 4th, Grishko V. Mitochondrial DNA damage is involved in apoptosis caused by pro-inflammatory cytokines in human OA chondrocytes. Osteoarthritis Cartilage. 2010; 18: 424–32.
  • Newsholme P, Curi R, Pithon Curi TC, Murphy CJ, Garcia C, Pires de Melo M. Glutamine metabolism by lymphocytes, macrophages, and neutrophils: its importance in health and disease. J Nutr Biochem. 1999; 10: 316–24.
  • Newsholme P. Why is L-glutamine metabolism important to cells of the immune system in health, postinjury, surgery or infection?. J Nutr. 2001;131 ( 9 Suppl): 2515S–22S.; discussion 2523S-4S.
  • Frost DV, Lish PM. Selenium in biology. Annu Rev Pharmacol. 1975; 15: 259–84.
  • Yamori Y, Taguchi T, Hamada A, Kunimasa K, Mori H, Mori M. Taurine in health and diseases: consistent evidence from experimental and epidemiological studies. J Biomed Sci. 2010; 1(17 Suppl): S6.
  • Amanzada A, Malik IA, Nischwitz M, Sultan S, Naz N, Ramadori G. Myeloperoxidase and elastase are only expressed by neutrophils in normal and in inflamed liver. Histochem Cell Biol. 2011; 135: 305–15.
  • Bunbury A, Potolicchio I, Maitra R, Santambrogio L. Functional analysis of monocyte MHC class II compartments. FASEB J. 2009; 23: 164–71.
  • Ray NJ, Jones AJ, Keen P. GABAB receptor modulation of the release of substance P from capsaicin-sensitive neurones in the rat trachea in vitro. Br J Pharmacol. 1991; 102: 801–4.
  • Chapman RW, Hey JA, Rizzo CA, Bolser DC. GABAB receptors in the lung. Trends Pharmacol Sci. 1993; 14: 26–9.
  • Minocha A, Galligan JJ. Excitatory and inhibitory responses mediated by GABAA and GABAB receptors in guinea pig distal colon. Eur J Pharmacol. 1993; 230: 187–93.
  • Ozdem SS, Sadan G, Usta C, Tasatargil A. Effect of experimental diabetes on GABA-mediated inhibition of neurally induced contractions in rat isolated trachea. Clin Exp Pharmacol Physiol. 2000; 27: 299–305.
  • Gentilini G, Franchi-Micheli S, Mugnai S, Bindi D, Zilletti L. GABA-mediated inhibition of the anaphylactic response in the guinea-pig trachea. Br J Pharmacol. 1995; 115: 389–94.
  • Ruiz de Valderas RM, Serrano MI, Serrano JS, Fernandez A. Effect of homotaurine in experimental analgesia tests. Gen Pharmacol. 1991; 22: 717–21.
  • Serrano I, Ruiz RM, Serrano JS, Fernandez A. GABAergic and cholinergic mediation in the antinociceptive action of homotaurine. Gen Pharmacol. 1992; 23: 421–6.
  • Silva MA, Cunha GM, Viana GS, Rao VS. Taurine modulates chemical nociception in mice. Braz J Med Biol Res. 1993; 26: 1319–24.
  • Serrano MI, Serrano JS, Guerrero MR, Fernandez A. Role of GABAA and GABAB receptors and peripheral cholinergic mechanisms in the antinociceptive action of taurine. Gen Pharmacol. 1994; 25: 1123–9.
  • Serrano MI, Serrano JS, Fernandez A, Asadi I, Serrano-Martino MC. GABAB receptors and opioid mechanisms involved in homotaurine-induced analgesia. Gen Pharmacol. 1998; 30: 411–5.
  • Cortijo J, Blesa S, Martinez-Losa M, Mata M, Seda E, Santangelo F, Morcillo EJ. Effects of taurine on pulmonary responses to antigen in sensitized Brown-Norway rats. Eur J Pharmacol. 2001; 431: 111–7.
  • Covarrubias J. Taurine and the lung: pharmacological intervention by aerosol route. Adv Exp Med Biol. 1994; 359: 413–7.
  • Fugelli K, Zachariassen KE. The distribution of taurine, gamma-aminobutyric acid and inorganic ions between plasma and erythrocytes in flounder (Platichthys flesus) at different plasma osmolalities. Comp Biochem Physiol A Comp Physiol. 1976; 55: 173–7.
  • Wapnir RA, Teichberg S. Regulation mechanisms of intestinal secretion: implications in nutrient absorption. J Nutr Biochem. 2002; 13: 190–199.
  • Farthing MJ. Novel agents for the control of secretory diarrhoea. Expert Opin Investig Drugs. 2004; 13: 777–85.
  • Lucas ML. Enterocyte chloride and water secretion into the small intestine after enterotoxin challenge: unifying hypothesis or intellectual dead end?. J Physiol Biochem. 2008; 64: 69–88.
  • Hartung HP, Toyka KV. Activation of macrophages by substance P: induction of oxidative burst and thromboxane release. Eur J Pharmacol. 1983; 89: 301–5.
  • Hartung HP, Wolters K, Toyka KV. Substance P: binding properties and studies on cellular responses in guinea pig macrophages. J Immunol. 1986; 136: 3856–63.
  • Peck R. Neuropeptides modulating macrophage function. Ann N Y Acad Sci. 1987; 496: 264–70.
  • Brunelleschi S, Vanni L, Ledda F, Giotti A, Maggi CA, Fantozzi R. Tachykinins activate guinea-pig alveolar macrophages: involvement of NK2 and NK1 receptors. Br J Pharmacol. 1990; 100: 417–20.
  • Boichot E, Lagente V, Paubert-Braquet M, Frossard N. Inhaled substance P induces activation of alveolar macrophages and increases airway responses in the guinea-pig. Neuropeptides. 1993; 25: 307–13.
  • Berman AS, Chancellor-Freeland C, Zhu G, Black PH. Substance P primes murine peritoneal macrophages for an augmented proinflammatory cytokine response to lipopolysaccharide. Neuroimmunomodulation. 1996; 3: 141–9.
  • Marriott I, Mason MJ, Elhofy A, Most KL. Substance P activates NF-kappaB independent of elevations in intracellular calcium in murine macrophages and dendritic cells. J Neuroimmunol. 2000; 102: 163–71.
  • Marriott I, Bost KL. Substance P receptor mediated macrophage responses. Adv Exp Med Biol. 2001; 493: 247–54.
  • Delgado AV, McManus AT, Chambers JP. Production of tumor necrosis factor-alpha, interleukin 1-beta, interleukin 2, and interleukin 6 by rat leukocyte subpopulations after exposure to substance P. Neuropeptides. 2003; 37: 355–61.
  • Weinstock JV. The role of substance P, hemokinin and their receptor in governing mucosal inflammation and granulomatous responses. Front Biosci. 2004; 9: 1936–43.
  • Bardelli C, Gunella G, Varsaldi F, Balbo P, Del Boca E, Bernardone IS, Amoruso A, Brunelleschi S. Expression of functional NK1 receptors in human alveolar macrophages: superoxide anion production, cytokine release and involvement of NF-kappaB pathway. Br J Pharmacol. 2005; 145: 385–96.
  • Koon HW, Pothoulakis C. Immunomodulatory properties of substance P: the gastrointestinal system as a model. Ann N Y Acad Sci. 2006; 1088: 23–40.
  • Serra MC, Bazzoni F, Della Bianca V, Greskowiak M, Rossi F. Activation of human neutrophils by substance P. Effect on oxidative metabolism, exocytosis, cytosolic Ca2 +  concentration and inositol phosphate formation. J Immunol. 1988; 141: 2118–24.
  • Perianin A, Snyderman R, Malfroy B. Substance P primes human neutrophil activation: a mechanism for neurological regulation of inflammation. Biochem Biophys Res Commun. 1989; 161: 520–4.
  • Brunelleschi S, Tarli S, Giotti A, Fantozzi R. Priming effects of mammalian tachykinins on human neutrophils. Life Sci. 1991; 48: PL1–5.
  • Kudlacz EM, Knippenberg RW. In vitro and in vivo effects of tachykinins on immune cell function in guinea pig airways. J Neuroimmunol. 1994; 50: 119–25.
  • Tanabe T, Otani H, Zeng XT, Mishima K, Ogawa R, Inagaki C. Inhibitory effects of calcitonin gene-related peptide on substance-P-induced superoxide production in human neutrophils. Eur J Pharmacol. 1996;314: 175–83. Erratum in: Eur J Pharmacol 1997;321:137-41.
  • Tanabe T, Otani H, Bao L, Mikami Y, Yasukura T, Ninomiya T, Ogawa R, Inagaki C. Intracellular signaling pathway of substance P-induced superoxide production in human neutrophils. Eur J Pharmacol. 1996; 299: 187–95.
  • Kontny E, Szczepańska K, Kowalczewski J, Kurowska M, Janicka I, Marcinkiewicz J, Maśliński W. The mechanism of taurine chloramine inhibition of cytokine (interleukin-6, interleukin-8) production by rheumatoid arthritis fibroblast-like synoviocytes. Arthritis Rheum. 2000; 43: 2169–77.
  • Barua M, Liu Y, Quinn MR. Taurine chloramine inhibits inducible nitric oxide synthase and TNF-alpha gene expression in activated alveolar macrophages: decreased NF-kappaB activation and IkappaB kinase activity. J Immunol. 2001; 167: 2275–81.
  • Liu Y, Quinn MR. Chemokine production by rat alveolar macrophages is inhibited by taurine chloramine. Immunol Lett. 2002; 80: 27–32.
  • Liu Y, Barua M, Serban V, Quinn MR. Production of inflammatory mediators by activated C6 cells is attenuated by taurine chloramine inhibition of NF-kappaB activation. Adv Exp Med Biol. 2003; 526: 365–72.
  • Quinn MR, Barua M, Liu Y, Serban V. Taurine chloramine inhibits production of inflammatory mediators and iNOS gene expression in alveolar macrophages; a tale of two pathways: part I, NF-kappaB signaling. Adv Exp Med Biol. 2003; 526: 341–8.
  • Schuller-Levis GB, Park E. Taurine: new implications for an old amino acid. FEMS Microbiol Lett. 2003; 226: 195–202.
  • Mainnemare A, Megarbane B, Soueidan A, Daniel A, Chapple IL. Hypochlorous acid and taurine-N-monochloramine in periodontal diseases. J Dent Res. 2004; 83: 823–31.
  • Kim JW, Kim C. Inhibition of LPS-induced NO production by taurine chloramine in macrophages is mediated though Ras-ERK-NF-kappaB. Biochem Pharmacol. 2005; 70: 1352–60.
  • Jackett PS, Aber VR, Lowrie DB. Virulence of Mycobacterium tuberculosis and susceptibility to peroxidative killing systems. J Gen Microbiol. 1978; 107: 273–8.
  • van Dalen CJ, Kettle AJ. Substrates and products of eosinophil peroxidase. Biochem J. 2001; 358: 233–9.
  • Klebanoff SJ, Kazazi F. Inactivation of human immunodeficiency virus type 1 by the amine oxidase-peroxidase system. J Clin Microbiol. 1995; 33: 2054–7.
  • Buys J, Wever R, van Stigt R, Ruitenberg EJ. The killing of newborn larvae of Trichinella spiralis by eosinophil peroxidase in vitro. Eur J Immunol. 1981; 11: 843–5.
  • Anderson WA, Kang YH, Mohla S. Mammalian endogenous peroxidases as cellular markers and as biosynthetic endpoints of hormone-mediated activity: viewpoint from cytochemistry. Prog Histochem Cytochem. 1979; 11: 1–27.
  • Krauskopf KB. Introduction to Geochemistry2. Ed. McGraw-Hill Book Company: Singapore, 1982
  • Koivistoinen P. Mineral Element Composition of Finnish Foods: N,K,Ca,Mg,P,S,Fe,Cu,Mn,Zn,Mo,Co,Ni,Cr,F,Se,Si,Rb,Al,B,Br,Hg,As,Cd,Pb and Ash. Acta Agriculturae Scandinavica. Supplementum 22. Stockholm 1980.
  • Helz GR, Hsu RY. Volatile chloro- and bromocarbons in coastal waters. Limnol Oceanogr. 1978; 23: 858–69.
  • Nagata M, Saito K. The roles of cysteinyl leukotrienes in eosinophilic inflammation of asthmatic airways. Int Arch Allergy Immunol. 2003; 1(131 Suppl): 7–10.
  • Mori M, Takaku Y, Kobayashi T, Hagiwara K, Kanazawa M, Nagata M. Eosinophil superoxide anion generation induced by adhesion molecules and leukotriene D4. Int Arch Allergy Immunol. 2009; 1(149 Suppl): 31–8.
  • Nakagome K, Nagata M. Pathogenesis of airway inflammation in bronchial asthma. Auris Nasus Larynx. 2011Feb 18. [Epub ahead of print].
  • Wang W, Hansbro PM, Foster PS, Yang M. An alternate STAT6-independent pathway promotes eosinophil influx into blood during allergic airway inflammation. PLoS One. 2011; 6: e17766.
  • Figtree GA, Liu CC, Bibert S, Hamilton EJ, Garcia A, White CN, Chia KK, Cornelius F, Geering K, Rasmussen HH. Reversible oxidative modification: a key mechanism of Na+-K+ pump regulation. Circ Res. 2009; 105: 185–93.
  • Szabó C, Módis K. Pathophysiological roles of peroxynitrite in circulatory shock. Shock. 2010; 1(34 Suppl): 4–14.
  • White CN, Liu CC, Garcia A, Hamilton EJ, Chia KK, Figtree GA, Rasmussen HH. Activation of cAMP-dependent signaling induces oxidative modification of the cardiac Na+-K+ pump and inhibits its activity. J Biol Chem. 2010; 285: 13712–20.
  • Ying J, Sharov V, Xu S, Jiang B, Gerrity R, Schöneich C, Cohen RA. Cysteine-674 oxidation and degradation of sarcoplasmic reticulum Ca2 +  ATPase in diabetic pig aorta. Free Radic Biol Med. 2008; 45: 756–62.
  • Lancel S, Qin F, Lennon SL, Zhang J, Tong X, Mazzini MJ, Kang YJ, Siwik DA, Cohen RA, Colucci WS. Oxidative posttranslational modifications mediate decreased SERCA activity and myocyte dysfunction in Galphaq-overexpressing mice. Circ Res. 2010; 107: 228–32.
  • Tang WH, Cheng WT, Kravtsov GM, Tong XY, Hou XY, Chung SK, Chung SS. Cardiac contractile dysfunction during acute hyperglycemia due to impairment of SERCA by polyol pathway-mediated oxidative stress. Am J Physiol Cell Physiol. 2010; 299: C643–53.
  • Rådmark O, Samuelsson B. 5-Lipoxygenase: mechanisms of regulation. J Lipid Res. 2009;50 Suppl: S40–5.
  • Manevich Y, Fisher AB. Peroxiredoxin 6, a 1-Cys peroxiredoxin, functions in antioxidant defense and lung phospholipid metabolism. Free Radic Biol Med. 2005; 38: 1422–32.
  • Fisher AB. Peroxiredoxin 6: A bifunctional enzyme with glutathione peroxidase and phospholipase A2 activities. Antioxid Redox Signal. 2010 Oct 4. [Epub ahead of print].
  • Monteiro G, Horta BB, Pimenta DC, Augusto O, Netto LE. Reduction of 1-Cys peroxiredoxins by ascorbate changes the thiol-specific antioxidant paradigm, revealing another function of vitamin C. Proc Natl Acad Sci U S A. 2007; 104: 4886–91.
  • Peshenko IV, Shichi H. Oxidation of active center cysteine of bovine 1-Cys peroxiredoxin to the cysteine sulfenic acid form by peroxide and peroxynitrite. Free Radic Biol Med. 2001; 31: 292–303.
  • Fujii T, Fujii J, Taniguchi N. Augmented expression of peroxiredoxin VI in rat lung and kidney after birth implies an antioxidative role. Eur J Biochem. 2001; 268: 218–25.
  • Liu G, Feinstein SI, Wang Y, Dodia C, Fisher D, Yu K, Ho YS, Fisher AB. Comparison of glutathione peroxidase 1 and peroxiredoxin 6 in protection against oxidative stress in the mouse lung. Free Radic Biol Med. 2010; 49: 1172–81.
  • Ding Y, Lu B, Chen D, Meng L, Shen Y, Chen S. Proteomic analysis of colonic mucosa in a rat model of irritable bowel syndrome. Proteomics. 2010; 10: 2620–30.
  • Naito Y, Takagi T, Okada H, Omatsu T, Mizushima K, Handa O, Kokura S, Ichikawa H, Fujiwake H, Yoshikawa T. Identification of inflammation-related proteins in a murine colitis model by 2D fluorescence difference gel electrophoresis and mass spectrometry. J Gastroenterol Hepatol. 2010; 1(25 Suppl): S144–8.
  • Morrison J, Knoll K, Hessner MJ, Liang M. Effect of high glucose on gene expression in mesangial cells: upregulation of the thiol pathway is an adaptational response. Physiol Genomics. 2004; 17: 271–82.
  • Wu J, Wang F, Gong Y, Li D, Sha J, Huang X, Han X. Proteomic analysis of changes induced by nonylphenol in Sprague-Dawley rat Sertoli cells. Chem Res Toxicol. 2009; 22: 668–75.
  • Leyens G, Verhaeghe B, Landtmeters M, Marchandise J, Knoops B, Donnay I. Peroxiredoxin 6 is upregulated in bovine oocytes and cumulus cells during in vitro maturation: role of intercellular communication. Biol Reprod. 2004; 71: 1646–51.
  • Martins-de-Souza D, Gattaz WF, Schmitt A, Novello JC, Marangoni S, Turck CW, Dias-Neto E. Proteome analysis of schizophrenia patients Wernicke's area reveals an energy metabolism dysregulation. BMC Psychiatry. 2009; 9: 17.
  • Sung JH, Cho EH, Kim MO, Koh PO. Identification of proteins differentially expressed by melatonin treatment in cerebral ischemic injury-a proteomics approach. J Pineal Res. 2009; 46: 300–6.
  • Srivastava AK. Assessment of salinity-induced antioxidative defense system of diazotrophic cyanobacterium Nostoc muscorum. J Microbiol Biotechnol. 2010; 20: 1506–12.
  • Elbaz A, Wei YY, Meng Q, Zheng Q, Yang ZM. Mercury-induced oxidative stress and impact on antioxidant enzymes in Chlamydomonas reinhardtii. Ecotoxicology. 2010; 19: 1285–93.
  • Sajitha Rajan S, Murugan K. Purification and kinetic characterization of the liverwort Pallavicinia lyelli (Hook.) S. Gray. cytosolic ascorbate peroxidase. Plant Physiol Biochem. 2010; 48: 758–63.
  • Kaul K, Lam KW, Fong D, Lok C, Berry M, Treble D. Ascorbate peroxidase in bovine retinal pigment epithelium and choroid. Curr Eye Res. 1988; 7: 675–9.
  • Wada N, Kinoshita S, Matsuo M, Amako K, Miyake C, Asada K. Purification and molecular properties of ascorbate peroxidase from bovine eye. Biochem Biophys Res Commun. 1998; 242: 256–61.
  • Mohr D, Bowry VW, Stocker R. Dietary supplementation with coenzyme Q10 results in increased levels of ubiquinol-10 within circulating lipoproteins and increased resistance of human low-density lipoprotein to the initiation of lipid peroxidation. Biochim Biophys Acta. 1992; 1126: 247–54.
  • Mohr D, Stocker R. Radical-mediated oxidation of isolated human very-low-density lipoprotein. Arterioscler Thromb. 1994; 14: 1186–92.
  • Tomasetti M, Littarru GP, Stocker R, Alleva R. Coenzyme Q10 enrichment decreases oxidative DNA damage in human lymphocytes. Free Radic Biol Med. 1999; 27: 1027–32.
  • Littarru GP, Tiano L. Bioenergetic and antioxidant properties of coenzyme Q10: recent developments. Mol Biotechnol. 2007; 37: 31–7.
  • Belardinelli R, Tiano L, Littarru GP. Oxidative stress, endothelial function and coenzyme Q10. Biofactors. 2008; 32: 129–33.
  • Kulakowski EC, Maturo J. Hypoglycemic properties of taurine: not mediated by enhanced insulin release. Biochem Pharmacol. 1984; 33: 2835–8.
  • Maturo J 3rd, Kulakowski EC. Insulin-like activity of taurine. Adv Exp Med Biol. 1987; 217: 217–26.
  • Maturo J, Kulakowski EC. Taurine binding to the purified insulin receptor. Biochem Pharmacol. 1988; 37: 3755–60.
  • Sugihara H. Nagasawa S, Okabe H. Experimentelle und klinische Untersuchungen über Taurin. [Experimental and clinical studies on taurine] [Article in German], Klin. Wochenschrift. 1936; 15: 751–756.
  • Christophersen OA. Fiskeproteinkonsentrat – et næringsmiddel for fremtiden? [Fish protein concentrate – a food for the future?] [Article in Norwegian]. Inter Medicos. 1977; 23: 53–67.
  • Carver JD. Dietary nucleotides: effects on the immune and gastrointestinal systems. Acta Paediatr Suppl. 1999; 88: 83–8.
  • Carver JD. Advances in nutritional modifications of infant formulas. Am J Clin Nutr. 2003 Jun; 77: 1550S–1554S.
  • Jacobsen JG, Smith LH. Biochemistry and physiology of taurine and taurine derivatives. Physiol Rev. 1968; 48: 424–511.
  • Fincham DA, Wolowyk MW, Young JD. Volume-sensitive taurine transport in fish erythrocytes. J Membr Biol. 1987; 96: 45–56.
  • Thoroed SM, Fugelli K. The Na+-independent taurine influx in flounder erythrocytes and its association with the volume regulatory taurine efflux. J Exp Biol. 1994; 186: 245–68.
  • Gaull GE, Wright CE, Tallan HH. Taurine in human lymphoblastoid cells: uptake and role in proliferation. Prog Clin Biol Res. 1983; 125: 297–303.
  • Nishio S, Negoro S, Hosokawa T, Hara H, Tanaka T, Deguchi Y, Ling J, Awata N, Azuma J, Aoike A, et al. The effect of taurine on age-related immune decline in mice: the effect of taurine on T cell and B cell proliferative response under costimulation with ionomycin and phorbol myristate acetate. Mech Ageing Dev. 1990; 52: 125–39.
  • Negoro S, Hara H. The effect of taurine on the age-related decline of the immune response in mice: the restorative effect on the T cell proliferative response to costimulation with ionomycin and phorbol myristate acetate. Adv Exp Med Biol. 1992; 315: 229–39.
  • Wang L, Zhao N, Zhang F, Yue W, Liang M. Effect of taurine on leucocyte function. Eur J Pharmacol. 2009; 616: 275–80.
  • Fazzino F, Obregón F, Lima L. Taurine and proliferation of lymphocytes in physically restrained rats. J Biomed Sci. 2010; 1(17 Suppl): S24.
  • Cubillos S, Urbina M, Lima L. Differential taurine effect on outgrowth from goldfish retinal ganglion cells after optic crush or axotomy. Influence of the optic tectum. Int J Dev Neurosci. 2000; 18: 843–53.
  • Cubillos S, Lima L. Taurine trophic modulation of goldfish retinal outgrowth and its interaction with the optic tectum. Amino Acids. 2006; 31: 325–31.
  • Hernández-Benítez R, Pasantes-Morales H, Saldaña IT, Ramos-Mandujano G. Taurine stimulates proliferation of mice embryonic cultured neural progenitor cells. J Neurosci Res. 2010; 88: 1673–81.
  • Michalk DV, Wingenfeld P, Licht C, Ugur T, Siar LF. The mechanisms of taurine mediated protection against cell damage induced by hypoxia and reoxygenation. Adv Exp Med Biol. 1996; 403: 223–32.
  • Schliess F, Häussinger D. Cell hydration and insulin signalling. Cell Physiol Biochem. 2000; 10: 403–8.
  • Schliess F, Häussinger D. Cell volume and insulin signaling. Int Rev Cytol. 2003; 225: 187–228.
  • vom Dahl S, Schliess F, Reissmann R, Görg B, Weiergräber O, Kocalkova M, Dombrowski F, Häussinger D. Involvement of integrins in osmosensing and signaling toward autophagic proteolysis in rat liver. J Biol Chem. 2003; 278: 27088–95.
  • Häussinger D. Osmosensing and osmosignaling in the liver. Wien Med Wochenschr. 2008; 158: 549–52.
  • Dogra C, Changotra H, Wedhas N, Qin X, Wergedal JE, Kumar A. TNF-related weak inducer of apoptosis (TWEAK) is a potent skeletal muscle-wasting cytokine. FASEB J. 2007; 21: 1857–69.
  • Judge AR, Koncarevic A, Hunter RB, Liou HC, Jackman RW, Kandarian SC. Role for IkappaBalpha, but not c-Rel, in skeletal muscle atrophy. Am J Physiol Cell Physiol. 2007; 292: C372–82.
  • Wu CL, Kandarian SC, Jackman RW. Identification of genes that elicit disuse muscle atrophy via the transcription factors p50 and Bcl-3. PLoS One. 2011; 6: e16171.
  • Hansen JM, Zhang H, Jones DP. Mitochondrial thioredoxin-2 has a key role in determining tumor necrosis factor-alpha-induced reactive oxygen species generation, NF-kappaB activation, and apoptosis. Toxicol Sci. 2006; 91: 643–50.
  • Yao H, Yang SR, Kode A, Rajendrasozhan S, Caito S, Adenuga D, Henry R, Edirisinghe I, Rahman I. Redox regulation of lung inflammation: role of NADPH oxidase and NF-kappaB signalling. Biochem Soc Trans. 2007; 35: 1151–5.
  • Rajendrasozhan S, Yang SR, Edirisinghe I, Yao H, Adenuga D, Rahman I. Deacetylases and NF-kappaB in redox regulation of cigarette smoke-induced lung inflammation: epigenetics in pathogenesis of COPD. Antioxid Redox Signal. 2008; 10: 799–811.
  • Hunter RB, Stevenson E, Koncarevic A, Mitchell-Felton H, Essig DA, Kandarian SC. Activation of an alternative NF-kappaB pathway in skeletal muscle during disuse atrophy. FASEB J. 2002; 16: 529–38.
  • Davies KJ. Degradation of oxidized proteins by the 20S proteasome. Biochimie. 2001; 83: 301–10.
  • Shringarpure R, Grune T, Davies KJ. Protein oxidation and 20S proteasome-dependent proteolysis in mammalian cells. Cell Mol Life Sci. 2001; 58: 1442–50.
  • Shringarpure R, Grune T, Mehlhase J, Davies KJ. Ubiquitin conjugation is not required for the degradation of oxidized proteins by proteasome. J Biol Chem. 2003; 278: 311–8.
  • Kurepa J, Smalle JA. To misfold or to lose structure?: Detection and degradation of oxidized proteins by the 20S proteasome. Plant Signal Behav. 2008; 3: 386–8.
  • McClung JM, Judge AR, Powers SK, Yan Z. p38 MAPK links oxidative stress to autophagy-related gene expression in cachectic muscle wasting. Am J Physiol Cell Physiol. 2010; 298: C542–9.
  • Zhang X, Tenner TE Jr, Lombardini JB. Inhibition of rat vascular smooth muscle cell proliferation by taurine and taurine analogues. Biochem Pharmacol. 1999; 57: 1331–9.
  • Ren K, Wang YC, Yang SJ. Effects of taurine on proliferation of rat cardiac fibroblast]. [Article in Chinese]. Yao Xue Xue Bao. 2008; 43: 591–5.
  • Murakami S, Sakurai T, Toda Y, Morito A, Sakono M, Fukuda N. Prevention of neointima formation by taurine ingestion after carotid balloon injury. Vascul Pharmacol. 2010; 53: 177–84.
  • Xie H, Yang B, Zhou XM, Song FL, Li JM, Zhou K, Hu W, Peng YQ, Tang SY, Yuan LQ, Xiong SY, Liao XB. L-carnitine and taurine synergistically inhibit the proliferation and osteoblastic differentiation of vascular smooth muscle cells. Acta Pharmacol Sin. 2010; 31: 289–96.
  • Leonarduzzi G, Scavazza A, Biasi F, Chiarpotto E, Camandola S, Vogel S, Dargel R, Poli G. The lipid peroxidation end product 4-hydroxy − 2,3-nonenal up-regulates transforming growth factor beta1 expression in the macrophage lineage: a link between oxidative injury and fibrosclerosis. FASEB J. 1997; 11: 851–7.
  • Cordain L, Eaton SB, Miller JB, Mann N, Hill K. The paradoxical nature of hunter-gatherer diets: meat-based, yet non-atherogenic. Eur J Clin Nutr. 2002; 1(56 Suppl): S42–52.
  • O'Keefe JH Jr, Cordain L. Cardiovascular disease resulting from a diet and lifestyle at odds with our Paleolithic genome: how to become a 21st-century hunter-gatherer. Mayo Clin Proc. 2004; 79: 101–8.
  • Frassetto LA, Schloetter M, Mietus-Synder M, Morris RC Jr, Sebastian A. Metabolic and physiologic improvements from consuming a Paleolithic, hunter-gatherer type diet. Eur J Clin Nutr. 2009; 63: 947–55.
  • Jönsson T, Granfeldt Y, Erlanson-Albertsson C, Ahrén B, Lindeberg S. A Paleolithic diet is more satiating per calorie than a Mediterranean-like diet in individuals with ischemic heart disease. Nutr Metab (Lond). 2010; 7: 85.
  • Ströhle A, Hahn A, Sebastian A. Latitude, local ecology, and hunter-gatherer dietary acid load: implications from evolutionary ecology. Am J Clin Nutr. 2010; 92: 940–5.
  • Stiner MC, Munro ND. On the evolution of diet and landscape during the Upper Paleolithic through Mesolithic at Franchthi Cave (Peloponnese, Greece). J Hum Evol. 2011; Mar 2. [Epub ahead of print].
  • Leakey RE, Lewin R. People of the Lake. Mankind & Its Beginnings. Avon Books., New York, 1979 ( First published 1978)
  • Eidlitz K. Food and emergency foods in the circumpolar area. Studia ethnographica Upsaliensis 32, University of Uppsala., Uppsala, 1969
  • Eidlitz K. Föda och nödföda – hur människan använde vildmarkens tillgångar [Food and emergency food – how humans were utilizing the resources of the wilderness] [Swedish book]. LT, Stockholm 1971
  • Dyerberg J. Coronary heart disease in Greenland Inuit: a paradox. Implications for western diet patterns. Arctic Med Res. 1989; 48: 47–54.
  • Dyerberg J, Schmidt EB. n-3 fatty acids and cardiovascular disease-observations generated by studies in Greenland Eskimos. Wien Klin Wochenschr. 1989; 101: 277–82.
  • Bigard AX. Risks of energy drinks in youths]. [Article in French]. Arch Pediatr. 2010; 17: 1625–31.
  • Kundurović Z, Sćepović M, Causević A, Mornjaković Z. Histochemical aspects and fine structural characteristics of thyreocytes in pinealectomized and melatonin treated rats prior to irradiation. Acta Med Croatica. 1991; 45: 347–55.
  • Mohan N, Sadeghi K, Reiter RJ, Meltz ML. The neurohormone melatonin inhibits cytokine, mitogen and ionizing radiation induced NF-kappa B. Biochem Mol Biol Int. 1995; 37: 1063–70.
  • Reiter RJ. The role of the neurohormone melatonin as a buffer against macromolecular oxidative damage. Neurochem Int. 1995; 27: 453–60.
  • Reiter RJ. Functional pleiotropy of the neurohormone melatonin: antioxidant protection and neuroendocrine regulation. Front Neuroendocrinol. 1995; 16: 383–415.
  • Reiter RJ, Melchiorri D, Sewerynek E, Poeggeler B, Barlow-Walden L, Chuang J, Ortiz GG, Acuña-Castroviejo D. A review of the evidence supporting melatonin's role as an antioxidant. J Pineal Res. 1995; 18: 1–11.
  • Vijayalaxmi, Reiter RJ, Meltz ML. Melatonin protects human blood lymphocytes from radiation-induced chromosome damage. Mutat Res. 1995;346: 23–31.
  • Vijayalaxmi., Reiter RJ, Sewerynek E, Poeggeler B, Leal BZ, Meltz ML. Marked reduction of radiation-induced micronuclei in human blood lymphocytes pretreated with melatonin. Radiat Res. 1995;143: 102–6.
  • Vijayalaxmi., Reiter RJ, Herman TS, Meltz ML. Melatonin and radioprotection from genetic damage: in vivo/in vitro studies with human volunteers. Mutat Res. 1996;371: 221–8.
  • Reiter R, Tang L, Garcia JJ, Muñoz-Hoyos A. Pharmacological actions of melatonin in oxygen radical pathophysiology. Life Sci. 1997; 60: 2255–71.
  • Reiter RJ, Guerrero JM, Garcia JJ, Acuña-Castroviejo D. Reactive oxygen intermediates, molecular damage, and aging. Relation to melatonin. Ann N Y Acad Sci. 1998; 854: 410–24.
  • Tan DX, Manchester LC, Reiter RJ, Plummer BF, Hardies LJ, Weintraub ST, Vijayalaxmi., Shepherd AM. A novel melatonin metabolite, cyclic 3-hydroxymelatonin: a biomarker of in vivo hydroxyl radical generation. Biochem Biophys Res Commun. 1998;253: 614–20.
  • Vijayalaxmi., Reiter RJ, Herman TS, Meltz ML. Melatonin reduces gamma radiation-induced primary DNA damage in human blood lymphocytes. Mutat Res. 1998;397: 203–8.
  • Vijayalaxmi, Reiter RJ, Meltz ML, Herman TS. Melatonin: possible mechanisms involved in its ‘radioprotective’ effect. Mutat Res. 1998;404: 187–9.
  • Badr FM, El Habit OH, Harraz MM. Radioprotective effect of melatonin assessed by measuring chromosomal damage in mitotic and meiotic cells. Mutat Res. 1999; 444: 367–72.
  • Reiter RJ. Oxidative damage to nuclear DNA: amelioration by melatonin. NEL Review. Neuro Endocrinol Lett. 1999; 20: 145–150.
  • Vijayalaxmi.,Meltz ML, Reiter RJ, Herman TS. Melatonin and protection from genetic damage in blood and bone marrow: whole-body irradiation studies in mice. J Pineal Res. 1999;27: 221–5.
  • Karbownik M, Reiter RJ. Antioxidative effects of melatonin in protection against cellular damage caused by ionizing radiation. Proc Soc Exp Biol Med. 2000; 225: 9–22.
  • Karbownik M, Reiter RJ, Qi W, Garcia JJ, Tan DX, Manchester LC, Vijayalaxmi. Protective effects of melatonin against oxidation of guanine bases in DNA and decreased microsomal membrane fluidity in rat liver induced by whole body ionizing radiation. Mol Cell Biochem. 2000;211: 137–44.
  • Kim JK, Lee CJ. Effect of exogenous melatonin on the ovarian follicles in gamma-irradiated mouse. Mutat Res. 2000; 449: 33–9.
  • Kim BC, Shon BS, Ryoo YW, Kim SP, Lee KS. Melatonin reduces X-ray irradiation-induced oxidative damages in cultured human skin fibroblasts. J Dermatol Sci. 2001; 26: 194–200.
  • Koc M, Buyukokuroglu ME, Taysi S. The effect of melatonin on peripheral blood cells during total body irradiation in rats. Biol Pharm Bull. 2002; 25: 656–7.
  • Vijayalaxmi., Thomas CR Jr, Reiter RJ, Herman TS. Melatonin: from basic research to cancer treatment clinics. J Clin Oncol. 2002;20: 2575–601.
  • Bonnefont-Rousselot D, Guilloz V, Lepage S, Bizard C, Duriez P, Lesieur D, Delattre J, Jore D, Gardès-Albert M. Protection of endogenous beta-carotene in LDL oxidized by oxygen free radicals in the presence of supraphysiological concentrations of melatonin. Redox Rep. 2003; 8: 95–104.
  • Koc M, Taysi S, Buyukokuroglu ME, Bakan N. Melatonin protects rat liver against irradiation-induced oxidative injury. J Radiat Res (Tokyo). 2003; 44: 211–5.
  • Koc M, Taysi S, Emin Buyukokuroglu M, Bakan N. The effect of melatonin against oxidative damage during total-body irradiation in rats. Radiat Res. 2003; 160: 251–5.
  • Sener G, Jahovic N, Tosun O, Atasoy BM, Yeğen BC. Melatonin ameliorates ionizing radiation-induced oxidative organ damage in rats. Life Sci. 2003; 74: 563–72.
  • Taysi S, Koc M, Büyükokuroğlu ME, Altinkaynak K, Sahin YN. Melatonin reduces lipid peroxidation and nitric oxide during irradiation-induced oxidative injury in the rat liver. J Pineal Res. 2003; 34: 173–7.
  • Yavuz MN, Yavuz AA, Ulku C, Sener M, Yaris E, Kosucu P, Karslioglu I. Protective effect of melatonin against fractionated irradiation-induced epiphyseal injury in a weanling rat model. J Pineal Res. 2003; 35: 288–94.
  • Weiss JF, Landauer MR. Protection against ionizing radiation by antioxidant nutrients and phytochemicals. Toxicology. 2003; 189: 1–20.
  • Erol FS, Topsakal C, Ozveren MF, Kaplan M, Ilhan N, Ozercan IH, Yildiz OG. Protective effects of melatonin and vitamin E in brain damage due to gamma radiation: an experimental study. Neurosurg Rev. 2004; 27: 65–9.
  • Oliınyk EV, Meshchyshen IF. Effect of melatonin and radiation on pro- and antioxidant state of the liver and blood of rats]. [Article in Ukrainian]. Ukr Biokhim Zh. 2004; 76: 144–7.
  • Reiter RJ, Tan DX, Gitto E, Sainz RM, Mayo JC, Leon J, Manchester LC, Vijayalaxmi., Kilic E, Kilic U. Pharmacological utility of melatonin in reducing oxidative cellular and molecular damage. Pol J Pharmacol. 2004;56: 159–70.
  • Undeger U, Giray B, Zorlu AF, Oge K, Baçaran N. Protective effects of melatonin on the ionizing radiation induced DNA damage in the rat brain. Exp Toxicol Pathol. 2004; 55: 379–84.
  • Sener G, Atasoy BM, Ersoy Y, Arbak S, Sengöz M, Yeğen BC. Melatonin protects against ionizing radiation-induced oxidative damage in corpus cavernosum and urinary bladder in rats. J Pineal Res. 2004; 37: 241–6.
  • Vijayalaxmi., Reiter RJ, Tan DX, Herman TS, Thomas CR Jr. Melatonin as a radioprotective agent: a review. Int J Radiat Oncol Biol Phys. 2004;59: 639–53.
  • Karslioglu I, Ertekin MV, Taysi S, Koçer I, Sezen O, Gepdiremen A, Koç M, Bakan N. Radioprotective effects of melatonin on radiation-induced cataract. J Radiat Res (Tokyo). 2005; 46: 277–82.
  • Monobe M, Hino M, Sumi M, Uzawa A, Hirayama R, Ando K, Kojima S. Protective effects of melatonin on gamma-ray induced intestinal damage. Int J Radiat Biol. 2005; 81: 855–60.
  • Alicelebić S, Mornjaković Z, Susko I, Cosović E, Beganović-Petrović A. The role of pineal gland and exogenous melatonin on the irradiation stress response of suprarenal gland. Bosn J Basic Med Sci. 2006; 6: 18–21.
  • Hussein MR, Abu-Dief EE, Abou El-Ghait AT, Adly MA, Abdelraheem MH. Melatonin and roentgen irradiation of the testis. Fertil Steril. 2006; 86: 750–2.
  • Hussein MR, Abu-Dief EE, Abou El-Ghait AT, Adly MA, Abdelraheem MH. Morphological evaluation of the radioprotective effects of melatonin against X-ray-induced early and acute testis damage in Albino rats: an animal model. Int J Exp Pathol. 2006; 87: 237–50.
  • Kopjar N, Miocić S, Ramić S, Milić M, Viculin T. Assessment of the radioprotective effects of amifostine and melatonin on human lymphocytes irradiated with gamma-rays in vitro. Arh Hig Rada Toksikol. 2006; 57: 155–63.
  • Kundurovic Z, Sofic E. The effects of exogenous melatonin on the morphology of thyrocytes in pinealectomized and irradiated rats. J Neural Transm. 2006; 113: 49–58.
  • Maurya DK, Devasagayam TP, Nair CK. Some novel approaches for radioprotection and the beneficial effect of natural products. Indian J Exp Biol. 2006; 44: 93–114.
  • Sharma S, Haldar C. Melatonin prevents X-ray irradiation induced oxidative damage in peripheral blood and spleen of the seasonally breeding rodent, Funambulus pennanti during reproductively active phase. Int J Radiat Biol. 2006; 82: 411–9.
  • Yilmaz S, Yilmaz E. Effects of melatonin and vitamin E on oxidative-antioxidative status in rats exposed to irradiation. Toxicology. 2006; 222: 1–7.
  • Zhou G, Kawata T, Furusawa Y, Aoki M, Hirayama R, Ando K, Ito H. Protective effects of melatonin against low- and high-LET irradiation. J Radiat Res (Tokyo). 2006; 47: 175–81.
  • El-Missiry MA, Fayed TA, El-Sawy MR, El-Sayed AA. Ameliorative effect of melatonin against gamma-irradiation-induced oxidative stress and tissue injury. Ecotoxicol Environ Saf. 2007; 66: 278–86.
  • Guney Y, Hicsonmez A, Uluoglu C, Guney HZ, Ozel Turkcu U, Take G, Yucel B, Caglar G, Bilgihan A, Erdogan D, Nalca Andrieu M, Kurtman C, Zengil H. Melatonin prevents inflammation and oxidative stress caused by abdominopelvic and total body irradiation of rat small intestine. Braz J Med Biol Res. 2007; 40: 1305–14.
  • Shirazi A, Ghobadi G, Ghazi-Khansari M. A radiobiological review on melatonin: a novel radioprotector. J Radiat Res (Tokyo). 2007; 48: 263–72.
  • Yildirim O, Comoğlu S, Yardimci S, Akmansu M, Bozkurt G, Avunduk MC. Melatonin treatment for prevention of oxidative stress: involving histopathological changes. Gen Physiol Biophys. 2007; 26: 126–32.
  • Hussein MR, Abu-Dief EE, Kamel E, Abou El-Ghait AT, Abdulwahed SR, Ahmad MH. Melatonin and roentgen irradiation-induced acute radiation enteritis in Albino rats: an animal model. Cell Biol Int. 2008; 32: 1353–61.
  • Reiter RJ, Korkmaz A. Clinical aspects of melatonin. Saudi Med J. 2008; 29: 1537–47.
  • Sharma S, Haldar C, Chaube SK. Effect of exogenous melatonin on X-ray induced cellular toxicity in lymphatic tissue of Indian tropical male squirrel, Funambulus pennanti. Int J Radiat Biol. 2008; 84: 363–74.
  • Taysi S, Memisogullari R, Koc M, Yazici AT, Aslankurt M, Gumustekin K, Al B, Ozabacigil F, Yilmaz A, Tahsin Ozder H. Melatonin reduces oxidative stress in the rat lens due to radiation-induced oxidative injury. Int J Radiat Biol. 2008; 84: 803–8.
  • Topkan E, Tufan H, Yavuz AA, Bacanli D, Onal C, Kosdak S, Yavuz MN. Comparison of the protective effects of melatonin and amifostine on radiation-induced epiphyseal injury. Int J Radiat Biol. 2008; 84: 796–802.
  • Yildirim O, Comoğlu S, Yardimci S, Akmansu M, Bozkurt G, Sürücü S. Preserving effects of melatonin on the levels of glutathione and malondialdehyde in rats exposed to irradiation. Gen Physiol Biophys. 2008; 27: 32–7.
  • Assayed ME, Abd El-Aty AM. Protection of rat chromosomes by melatonin against gamma radiation-induced damage. Mutat Res. 2009; 677: 14–20.
  • Hamada N. Recent insights into the biological action of heavy-ion radiation. J Radiat Res (Tokyo). 2009; 50: 1–9.
  • Jang SS, Kim WD, Park WY. Melatonin exerts differential actions on X-ray radiation-induced apoptosis in normal mice splenocytes and Jurkat leukemia cells. J Pineal Res. 2009; 47: 147–55.
  • Take G, Erdogan D, Helvacioglu F, Göktas G, Ozbey G, Uluoglu C, Yücel B, Guney Y, Hicsonmez A, Ozkan S. Effect of melatonin and time of administration on irradiation-induced damage to rat testes. Braz J Med Biol Res. 2009; 42: 621–8.
  • Sharma S, Haldar C, Chaube SK, Laxmi T, Singh SS. Long-term melatonin administration attenuates low-LET gamma-radiation-induced lymphatic tissue injury during the reproductively active and inactive phases of Indian palm squirrels (Funambulus pennanti). Br J Radiol. 2010; 83: 137–51.
  • Cekan E, Slanina P, Bergman K, Tribukait B. Effects of dietary supplementation with selenomethionine on the teratogenic effect of ionizing radiation in mice. Acta Radiol Oncol. 1985; 24: 459–63.
  • Cekan E, Tribukait B, Vokal-Borek H. Protective effect of selenium against ionizing radiation-induced malformations in mice. Acta Radiol Oncol. 1985; 24: 267–71.
  • Borek C, Ong A, Mason H, Donahue L, Biaglow JE. Selenium and vitamin E inhibit radiogenic and chemically induced transformation in vitro via different mechanisms. Proc Natl Acad Sci U S A. 1986; 83: 1490–4.
  • Hall EJ, Hei TK. Modulating factors in the expression of radiation-induced oncogenic transformation. Environ Health Perspect. 1990; 88: 149–55.
  • Knizhnikov VA, Komleva VA, Tutel'ian VA, Novoselova GP, Golubkina NA, Trushina EN, Kumpulaínen I, Edelman K. The effect of an increased dietary intake of organic selenium on the resistance of rats to ionizing radiation, aflatoxin B1 and infection]. [Article in Russian]. Vopr Pitan. 1991;(4): 52–5.
  • Weiss JF, Srinivasan V, Kumar KS, Landauer MR. Radioprotection by metals: selenium. Adv Space Res. 1992; 12: 223–31.
  • Sun E, Xu H, Liu Q, Zhou J, Zuo P, Wang J. Effect of selenium in recovery of immunity damaged by H2O2 and 60Co radiation. Biol Trace Elem Res. 1995; 48: 239–50.
  • Rodemann HP, Hehr T, Bamberg M. Relevance of the radioprotective effect of sodium selenite]. [Article in German]. Med Klin (Munich). 1999; 3(94 Suppl): 39–41.
  • Schleicher UM, Lopez Cotarelo C, Andreopoulos D, Handt S, Ammon J. Radioprotection of human endothelial cells by sodium selenite]. [Article in German]. Med Klin (Munich). 1999; 3(94 Suppl): 35–8.
  • Mutlu-Türkoğlu U, Erbil Y, Oztezcan S, Olgaç V, Toker G, Uysal M. The effect of selenium and/or vitamin E treatments on radiation-induced intestinal injury in rats. Life Sci. 2000; 66: 1905–13.
  • Yanardağ R, Bolkent S, Kizir A. Protective effects of DL-alpha-tocopherol acetate and sodium selenate on the liver of rats exposed to gamma radiation. Biol Trace Elem Res. 2001; 83: 263–73.
  • Micke O, Bruns F, Mücke R, Schäfer U, Glatzel M, DeVries AF, Schönekaes K, Kisters K, Büntzel J. Selenium in the treatment of radiation-associated secondary lymphedema. Int J Radiat Oncol Biol Phys. 2003; 56: 40–9.
  • Sagowski C, Wenzel S, Tesche S, Jenicke L, Kehrl W, Roeser K, Metternich FU. Sodium selenite reduces acute radiogenic damage of the rat parotid glands during fractionated irradiation]. [Article in German]. HNO. 2004; 52: 1067–75.
  • Sagowski C, Wenzel S, Jenicke L, Metternich FU, Jaehne M. Sodium selenite is a potent radioprotector of the salivary glands of the rat: acute effects on the morphology and parenchymal function during fractioned irradiation. Eur Arch Otorhinolaryngol. 2005; 262: 459–64.
  • Gehrisch A, Dörr W. Effects of systemic or topical administration of sodium selenite on early radiation effects in mouse oral mucosa. Strahlenther Onkol. 2007; 183: 36–42.
  • Pontual ML, Tuji FM, Barros SP, Bóscolo FN, Novaes PD, de Almeida SM. Ultrastructural evaluation of the radioprotective effect of sodium selenite on submandibular glands in rats. J Appl Oral Sci. 2007; 15: 162–8.
  • Baliga MS, Diwadkar-Navsariwala V, Koh T, Fayad R, Fantuzzi G, Diamond AM. Selenoprotein deficiency enhances radiation-induced micronuclei formation. Mol Nutr Food Res. 2008; 52: 1300–4.
  • Margulies BS, Damron TA, Allen MJ. The differential effects of the radioprotectant drugs amifostine and sodium selenite treatment in combination with radiation therapy on constituent bone cells, Ewing's sarcoma of bone tumor cells, and rhabdomyosarcoma tumor cells in vitro. J Orthop Res. 2008; 26: 1512–9.
  • Micke O, Schomburg L, Buentzel J, Kisters K, Muecke R. Selenium in oncology: from chemistry to clinics. Molecules. 2009; 14: 3975–88.
  • Rocha AS, Ramos-Perez FM, Bóscolo FN, Manzi FR, Cchicarelo M, Almeida SM. Effect of sodium selenite on bone repair in tibiae of irradiated rats. Braz Dent J. 2009; 20: 186–90.
  • Gençel O, Naziroglu M, Celik O, Yalman K, Bayram D. Selenium and vitamin E modulates radiation-induced liver toxicity in pregnant and nonpregnant rat: effects of colemanite and hematite shielding. Biol Trace Elem Res. 2010; 135: 253–63.
  • Muecke R, Schomburg L, Buentzel J, Kisters K, Micke O. German Working Group Trace Elements and Electrolytes in Oncology. Selenium or no selenium-that is the question in tumor patients: a new controversy. Integr Cancer Ther. 2010; 9: 136–41.
  • Tuji FM, Pontual ML, Barros SP, Almeida SM, Bóscolo FN. Ultrastructural assessment of the radioprotective effects of sodium selenite on parotid glands in rats. J Oral Sci. 2010; 52: 369–75.
  • Dethmers JK, Meister A. Glutathione export by human lymphoid cells: depletion of glutathione by inhibition of its synthesis decreases export and increases sensitivity to irradiation. Proc Natl Acad Sci U S A. 1981; 78: 7492–6.
  • Jensen GL, Meister A. Radioprotection of human lymphoid cells by exogenously supplied glutathione is mediated by gamma-glutamyl transpeptidase. Proc Natl Acad Sci U S A. 1983; 80: 4714–7.
  • Wellner VP, Anderson ME, Puri RN, Jensen GL, Meister A. Radioprotection by glutathione ester: transport of glutathione ester into human lymphoid cells and fibroblasts. Proc Natl Acad Sci U S A. 1984; 81: 4732–5.
  • Vos O, Roos-Verhey WS. Radioprotection by glutathione esters and cysteamine in normal and glutathione-depleted mammalian cells. Int J Radiat Biol Relat Stud Phys Chem Med. 1988; 53: 273–81.
  • Shaheen AA, Hassan SM. Radioprotection of whole-body gamma-irradiation-induced alteration in some haematological parameters by cysteine, vitamin E and their combination in rats. Strahlenther Onkol. 1991; 167: 498–501.
  • Sridharan S, Shyamaladevi CS. Protective effect of N-acetylcysteine against gamma ray induced damages in rats-biochemical evaluations. Indian J Exp Biol. 2002; 40: 181–6.
  • Neal R, Matthews RH, Lutz P, Ercal N. Antioxidant role of N-acetyl cysteine isomers following high dose irradiation. Free Radic Biol Med. 2003; 34: 689–95.
  • Liu Y, Zhang H, Zhang L, Zhou Q, Wang X, Long J, Dong T, Zhao W. Antioxidant N-acetylcysteine attenuates the acute liver injury caused by X-ray in mice. Eur J Pharmacol. 2007; 575: 142–8.
  • Kilciksiz S, Demirel C, Erdal N, Gürgül S, Tamer L, Ayaz L, Ors Y. The effect of N-acetylcysteine on biomarkers for radiation-induced oxidative damage in a rat model. Acta Med Okayama. 2008; 62: 403–9.
  • Mansour HH, Hafez HF, Fahmy NM, Hanafi N. Protective effect of N-acetylcysteine against radiation induced DNA damage and hepatic toxicity in rats. Biochem Pharmacol. 2008; 75: 773–80.
  • Demirel C, Kilçiksiz S, Ay OI, Gürgül S, Ay ME, Erdal N. Effect of N-acetylcysteine on radiation-induced genotoxicity and cytotoxicity in rat bone marrow. J Radiat Res (Tokyo). 2009; 50: 43–50.
  • Tiwari P, Kumar A, Balakrishnan S, Kushwaha HS, Mishra KP. Radiation-induced micronucleus formation and DNA damage in human lymphocytes and their prevention by antioxidant thiols. Mutat Res. 2009; 676: 62–8.
  • Hartman PE, Hartman Z, Citardi MJ. Ergothioneine, histidine, and two naturally occurring histidine dipeptides as radioprotectors against gamma-irradiation inactivation of bacteriophages T4 and P22. Radiat Res. 1988; 114: 319–30.
  • Guney Y, Turkcu UO, Hicsonmez A, Andrieu MN, Guney HZ, Bilgihan A, Kurtman C. Carnosine may reduce lung injury caused by radiation therapy. Med Hypotheses. 2006; 66: 957–9.
  • Altas E, Ertekin MV, Gundogdu C, Demirci E. L-carnitine reduces cochlear damage induced by gamma irradiation in Guinea pigs. Ann Clin Lab Sci. 2006; 36: 312–8.
  • Mansour HH. Protective role of carnitine ester against radiation-induced oxidative stress in rats. Pharmacol Res. 2006; 54: 165–71.
  • Uçüncü H, Ertekin MV, Yörük O, Sezen O, Ozkan A, Erdoğan F, Kiziltunç A, Gündoğdu C. Vitamin E and L-carnitine, separately or in combination, in the prevention of radiation-induced oral mucositis and myelosuppression: a controlled study in a rat model. J Radiat Res (Tokyo). 2006; 47: 91–102.
  • Kocer I, Taysi S, Ertekin MV, Karslioglu I, Gepdiremen A, Sezen O, Serifoglu K. The effect of L-carnitine in the prevention of ionizing radiation-induced cataracts: a rat model. Graefes Arch Clin Exp Ophthalmol. 2007; 245: 588–94.
  • Sezen O, Ertekin MV, Demircan B, Karslioğlu I, Erdoğan F, Koçer I, Calik I, Gepdiremen A. Vitamin E and L-carnitine, separately or in combination, in the prevention of radiation-induced brain and retinal damages. Neurosurg Rev. 2008;31:205-13; discussion 213.
  • Caloglu M, Yurut-Caloglu V, Durmus-Altun G, Oz-Puyan F, Ustun F, Cosar-Alas R, Saynak M, Parlar S, Turan FN, Uzal C. Histopathological and scintigraphic comparisons of the protective effects of L-carnitine and amifostine against radiation-induced late renal toxicity in rats. Clin Exp Pharmacol Physiol. 2009; 36: 523–30.
  • Topcu-Tarladacalisir Y, Kanter M, Uzal MC. Role of L-carnitine in the prevention of seminiferous tubules damage induced by gamma radiation: a light and electron microscopic study. Arch Toxicol. 2009; 83: 735–46.
  • Kanter M, Topcu-Tarladacalisir Y, Parlar S. Antiapoptotic effect of L-carnitine on testicular irradiation in rats. J Mol Histol. 2010; 41: 121–8.
  • Christophersen OA. Radiation biochemistry. In: Brune D, Hellborg R, Persson BRR, Pääkkönen. Radiation at Home, Outdoors and in the Workplace. Scandinavian Science Publishers: Oslo, 2001, pp. 52–61.
  • Kohen R, Yamamoto Y, Cundy KC, Ames BN. Antioxidant activity of carnosine, homocarnosine, and anserine present in muscle and brain. Proc Natl Acad Sci U S A. 1988; 85: 3175–9.
  • Aruoma OI, Laughton MJ, Halliwell B. Carnosine, homocarnosine and anserine: could they act as antioxidants in vivo?. Biochem J. 1989; 264: 863–9.
  • Babizhayev MA, Seguin MC, Gueyne J, Evstigneeva RP, Ageyeva EA, Zheltukhina GA. L-carnosine (beta-alanyl-L-histidine) and carcinine (beta-alanylhistamine) act as natural antioxidants with hydroxyl-radical-scavenging and lipid-peroxidase activities. Biochem J. 1994; 304: 509–16.
  • Kang JH, Kim KS, Choi SY, Kwon HY, Won MH, Kang TC. Carnosine and related dipeptides protect human ceruloplasmin against peroxyl radical-mediated modification. Mol Cells. 2002; 13: 498–502.
  • Kang JH, Kim KS, Choi SY, Kwon HY, Won MH, Kang TC. Protective effects of carnosine, homocarnosine and anserine against peroxyl radical-mediated Cu,Zn-superoxide dismutase modification. Biochim Biophys Acta. 2002; 1570: 89–96.
  • Kang JH, Kim KS, Choi SY, Kwon HY, Won MH, Kang TC. Protection by carnosine-related dipeptides against hydrogen peroxide-mediated ceruloplasmin modification. Mol Cells. 2002; 13: 107–12.
  • Hipkiss AR. Carnosine and its possible roles in nutrition and health. Adv Food Nutr Res. 2009; 57: 87–154.
  • Babizhayev MA, Yegorov YE. Reactive oxygen species and the aging eye: Specific role of metabolically active mitochondria in maintaining lens function and in the initiation of the oxidation-induced maturity onset cataract-a novel platform of mitochondria-targeted antioxidants with broad therapeutic potential for redox regulation and detoxification of oxidants in eye diseases. Am J Ther. 2010Oct 22. [Epub ahead of print].
  • Halliwell B, Gutteridge JMC. Free Radicals in Biology and MedicineFourth Edition. Oxford University Press. 2007
  • Fontana M, Pinnen F, Lucente G, Pecci L. Prevention of peroxynitrite-dependent damage by carnosine and related sulphonamido pseudodipeptides. Cell Mol Life Sci. 2002; 59: 546–51.
  • Yanai N, Shiotani S, Hagiwara S, Nabetani H, Nakajima M. Antioxidant combination inhibits reactive oxygen species mediated damage. Biosci Biotechnol Biochem. 2008; 72: 3100–6.
  • Nicoletti VG, Santoro AM, Grasso G, Vagliasindi LI, Giuffrida ML, Cuppari C, Purrello VS, Stella AM, Rizzarelli E. Carnosine interaction with nitric oxide and astroglial cell protection. J Neurosci Res. 2007; 85: 2239–45.
  • Babizhayev MA, Deyev AI. Management of the virulent influenza virus infection by oral formulation of nonhydrolized carnosine and isopeptide of carnosine attenuating proinflammatory cytokine-induced nitric oxide production. Am J Ther. 2010. Sep 14. [Epub ahead of print].
  • Tamura EK, Silva CL, Markus RP. Melatonin reduces nitric oxide synthase activity in rat hypothalamus. J Pineal Res. 2006; 41: 267–74.
  • Pozo D, Reiter RJ, Calvo JR, Guerrero JM. Inhibition of cerebellar nitric oxide synthase and cyclic GMP production by melatonin via complex formation with calmodulin. J Cell Biochem. 1997; 65: 430–42.
  • León J, Macías M, Escames G, Camacho E, Khaldy H, Martín M, Espinosa A, Gallo MA, Acuña-Castroviejo D. Structure-related inhibition of calmodulin-dependent neuronal nitric-oxide synthase activity by melatonin and synthetic kynurenines. Mol Pharmacol. 2000; 58: 967–75.
  • Sáenz DA, Turjanski AG, Sacca GB, Marti M, Doctorovich F, Sarmiento MI, Estrin DA, Rosenstein RE. Physiological concentrations of melatonin inhibit the nitridergic pathway in the Syrian hamster retina. J Pineal Res. 2002; 33: 31–6.
  • Tamura EK, Silva CL, Markus RP. Melatonin inhibits endothelial nitric oxide production in vitro. J Pineal Res. 2006; 41: 267–74.
  • Ouyang H, Vogel HJ. Melatonin and serotonin interactions with calmodulin: NMR, spectroscopic and biochemical studies. Biochim Biophys Acta. 1998; 1383: 37–47.
  • Fukunaga K, Horikawa K, Shibata S, Takeuchi Y, Miyamoto E. Ca2 + /calmodulin-dependent protein kinase II-dependent long-term potentiation in the rat suprachiasmatic nucleus and its inhibition by melatonin. J Neurosci Res. 2002; 70: 799–807.
  • Douki T, Cadet J. Peroxynitrite mediated oxidation of purine bases of nucleosides and isolated DNA. Free Radic Res. 1996; 24: 369–80.
  • Sodum RS, Fiala ES. Analysis of peroxynitrite reactions with guanine, xanthine, and adenine nucleosides by high-pressure liquid chromatography with electrochemical detection: C8-nitration and -oxidation. Chem Res Toxicol. 2001; 14: 438–50.
  • Cao Z, Li Y. Potent inhibition of peroxynitrite-induced DNA strand breakage by ethanol: possible implications for ethanol-mediated cardiovascular protection. Pharmacol Res. 2004; 50: 13–9.
  • Yu H, Venkatarangan L, Wishnok JS, Tannenbaum SR. Quantitation of four guanine oxidation products from reaction of DNA with varying doses of peroxynitrite. Chem Res Toxicol. 2005; 18: 1849–57.
  • Jia Z, Zhu H, Vitto MJ, Misra BR, Li Y, Misra HP. Alpha-lipoic acid potently inhibits peroxynitrite-mediated DNA strand breakage and hydroxyl radical formation: implications for the neuroprotective effects of alpha-lipoic acid. Mol Cell Biochem. 2009; 323: 131–8.
  • Chen W, Jia Z, Zhu H, Zhou K, Li Y, Misra HP. Ethyl pyruvate inhibits peroxynitrite-induced DNA damage and hydroxyl radical generation: implications for neuroprotection. Neurochem Res. 2010; 35: 336–42.
  • Kjosbakken J, Larsen H. The non-protein nitrogenous compounds of fish used in meal production, with special reference to capelin (Mallotus villosus). Fiskeridirektoratets Skrifter. Serie Ernæring. 1981; 2: 7–24.
  • Abe H, Dobson GP, Hoeger U, Parkhouse WS. Role of histidine-related compounds to intracellular buffering in fish skeletal muscle. Am J Physiol. 1985; 249: R449–54.
  • Suzuki T, Hirano T, Suyama M. Free imidazole compounds in white and dark muscles of migratory marine fish. Comp Biochem Physiol B. 1987; 87: 615–9.
  • Okuma E, Abe H. Major buffering constituents in animal muscle. Comp Biochem Physiol Comp Physiol. 1992; 102: 37–41.
  • Bragadóttir M. Endogenous antioxidants in fish. A literature review submitted in partial fulfilment of the requirements for the degree of MASTER OF SCIENCE in food science. Department of Food Science. University of Iceland. Reykjavik 2001. -.
  • Bragadóttir M, Reichert J, Jónsdóttir R, Ólafsdóttir G. Characterisation and antioxidant properties of aqueous extracts from capelin (Mallotus villosus). Verkefnaskýrsla Rf 39-06. Rannsóknastofnun fiskidnadarins/Icelandic Fisheries Laboratories. Desember 2006.
  • Blount BC, Mack MM, Wehr CM, MacGregor JT, Hiatt RA, Wang G, Wickramasinghe SN, Everson RB, Ames BN. Folate deficiency causes uracil misincorporation into human DNA and chromosome breakage: implications for cancer and neuronal damage. Proc Natl Acad Sci U S A. 1997; 94: 3290–5.
  • Li L, Connor EE, Berger SH, Wyatt MD. Determination of apoptosis, uracil incorporation, DNA strand breaks, and sister chromatid exchanges under conditions of thymidylate deprivation in a model of BER deficiency. Biochem Pharmacol. 2005; 70: 1458–68.
  • Berger SH, Pittman DL, Wyatt MD. Uracil in DNA: consequences for carcinogenesis and chemotherapy. Biochem Pharmacol. 2008; 76: 697–706.
  • Fenech M. Folate, DNA damage and the aging brain. Mech Ageing Dev. 2010; 131: 236–41.
  • Fenech M. The role of folic acid and vitamin B12 in genomic stability of human cells. Mutat Res. 2001; 475: 57–67.
  • Choi SW, Friso S, Ghandour H, Bagley PJ, Selhub J, Mason JB. Vitamin B-12 deficiency induces anomalies of base substitution and methylation in the DNA of rat colonic epithelium. J Nutr. 2004; 134: 750–5.
  • Kapiszewska M, Kalemba M, Wojciech U, Milewicz T. Uracil misincorporation into DNA of leukocytes of young women with positive folate balance depends on plasma vitamin B12 concentrations and methylenetetrahydrofolate reductase polymorphisms. A pilot study. J Nutr Biochem. 2005; 16: 467–78.
  • Hambidge KM, Casey CE, Krebs NF. Zinc. In: Mertz W. Trace Elements in Human and Animal Nutrition – Fifth Edition. Vol. 2. New York: Academic Press.: 1986, pp. 1–137.
  • Christophersen OA, Haug A, Steinnes E. Deforestation, mineral nutrient depletion in the soil and HIV disease. Science without borders. Transactions of the International Academy of Science H&E. Special Edition International Conference Oslo 2009. Innsbruck, SWB, 2010, pp. 26–34.
  • Gromer S, Arscott LD, Williams CH Jr, Schirmer RH, Becker K. Human placenta thioredoxin reductase. Isolation of the selenoenzyme, steady state kinetics, and inhibition by therapeutic gold compounds. J Biol Chem. 1998; 273: 20096–101.
  • Soldatenkov VA, Smulson M. Poly(ADP-ribose) polymerase in DNA damage-response pathway: implications for radiation oncology. Int J Cancer. 2000; 90: 59–67.
  • Soldatenkov VA, Potaman VN. DNA-binding properties of poly(ADP-ribose) polymerase: a target for anticancer therapy. Curr Drug Targets. 2004; 5: 357–65.
  • Lapucci A, Pittelli M, Rapizzi E, Felici R, Moroni F, Chiarugi A. Poly(ADP-ribose) polymerase-1 is a nuclear epigenetic regulator of mitochondrial DNA repair and transcription. Mol Pharmacol. 2011 Mar 11. [Epub ahead of print].
  • Rawling JM, Jackson TM, Driscoll ER, Kirkland JB. Dietary niacin deficiency lowers tissue poly(ADP-ribose) and NAD+ concentrations in Fischer-344 rats. J Nutr. 1994; 124: 1597–603.
  • Spronck JC, Kirkland JB. Niacin deficiency increases spontaneous and etoposide-induced chromosomal instability in rat bone marrow cells in vivo. Mutat Res. 2002; 508: 83–97.
  • Kostecki LM, Thomas M, Linford G, Lizotte M, Toxopeus L, Bartleman AP, Kirkland JB. Niacin deficiency delays DNA excision repair and increases spontaneous and nitrosourea-induced chromosomal instability in rat bone marrow. Mutat Res. 2007; 625: 50–61.
  • Yu TW, Anderson D. Reactive oxygen species-induced DNA damage and its modification: a chemical investigation. Mutat Res. 1997; 379: 201–10.
  • Brunton LD, Lazo JS, Parker KL. Goodman & Gilman's The Pharmacological Basis of Therapeutics. Eleventh edition. McGraw-Hill: New York, 2006
  • Mungunsukh O, Griffin AJ, Lee YH, Day RM. Bleomycin induces the extrinsic apoptotic pathway in pulmonary endothelial cells. Am J Physiol Lung Cell Mol Physiol. 2010 Feb 12. [Epub ahead of print].
  • Yong LC, Petersen MR. High dietary niacin intake is associated with decreased chromosome translocation frequency in airline pilots. Br J Nutr. 2011; 105: 496–505.
  • Ames BN. Micronutrient deficiencies. A major cause of DNA damage. Ann N Y Acad Sci. 1999; 889: 87–106.
  • Maestroni GJ. kappa-Opioid receptors in marrow stroma mediate the hematopoietic effects of melatonin-induced opioid cytokines. Ann N Y Acad Sci. 1998; 840: 411–9.
  • Maestroni GJ, Zammaretti F, Pedrinis E. Hematopoietic effect of melatonin involvement of type 1 kappa-opioid receptor on bone marrow macrophages and interleukin-1. J Pineal Res. 1999; 27: 145–53.
  • Molinero P, Soutto M, Benot S, Hmadcha A, Guerrero JM. Melatonin is responsible for the nocturnal increase observed in serum and thymus of thymosin alpha1 and thymulin concentrations: observations in rats and humans. J Neuroimmunol. 2000; 103: 180–8.
  • Arias J, Melean E, Valero N, Pons H, Chacín-Bonilla L, Larreal Y, Bonilla E. Effect of melatonin on lymphocyte proliferation and production of interleukin-2 (IL-2) and interleukin-1 beta (IL-1 beta) in mice splenocytes] [Article in Spanish]. Invest Clin. 2003; 44: 41–50.
  • Hoijman E, Rocha Viegas L, Keller Sarmiento MI, Rosenstein RE, Pecci A. Involvement of Bax protein in the prevention of glucocorticoid-induced thymocytes apoptosis by melatonin. Endocrinology. 2004; 145: 418–25.
  • Miller SC, Pandi-Perumal SR, Esquifino AI, Cardinali DP, Maestroni GJ. The role of melatonin in immuno-enhancement: potential application in cancer. Int J Exp Pathol. 2006;87: 81–7. Erratum in: Int J Exp Pathol. 2006;87:251. Pandi, Perumal SR [corrected to Pandi-Perumal, SR].
  • Presman DM, Hoijman E, Ceballos NR, Galigniana MD, Pecci A. Melatonin inhibits glucocorticoid receptor nuclear translocation in mouse thymocytes. Endocrinology. 2006; 147: 5452–9.
  • Batmanabane M. Melatonin is responsible for the nocturnal increase observed in serum and thymus of alpha1-thymosin and thymulin concentrations: observations in rats and humans. J Neuroimmunol. 2007; 183: 239; author reply 240.
  • Cardinali DP, Esquifino AI, Srinivasan V, Pandi-Perumal SR. Melatonin and the immune system in aging. Neuroimmunomodulation. 2008; 15: 272–8.
  • Srinivasan V, Spence DW, Trakht I, Pandi-Perumal SR, Cardinali DP, Maestroni GJ. Immunomodulation by melatonin: its significance for seasonally occurring diseases. Neuroimmunomodulation. 2008; 15: 93–101.
  • Zhou W, Wang P, Tao L. Effect of melatonin on proliferation of neonatal cord blood mononuclear cells. World J Pediatr. 2009; 5: 300–3.
  • Viviani S, Negretti E, Orazi A, Sozzi G, Santoro A, Lissoni P, Esposti G, Fraschini F. Preliminary studies on melatonin in the treatment of myelodysplastic syndromes following cancer chemotherapy. J Pineal Res. 1990; 8: 347–54.
  • Bregani ER, Lissoni P, Rossini F, Barni S, Tancini G, Brivio F, Conti A, Maestroni GJ. Prevention of interleukin-2-induced thrombocytopenia during the immunotherapy of cancer by a concomitant administration of the pineal hormone melatonin. Recenti Prog Med. 1995; 86: 231–3.
  • Lissoni P, Barni S, Brivio F, Rossini F, Fumagalli L, Tancini G. Treatment of cancer-related thrombocytopenia by low-dose subcutaneous interleukin-2 plus the pineal hormone melatonin: a biological phase II study. J Biol Regul Homeost Agents. 1995; 9: 52–4.
  • Lissoni P, Barni S, Brivio F, Rossini F, Fumagalli L, Ardizzoia A, Tancini G. A biological study on the efficacy of low-dose subcutaneous interleukin-2 plus melatonin in the treatment of cancer-related thrombocytopenia. Oncology. 1995; 52: 360–2.
  • Lissoni P, Tancini G, Barni S, Paolorossi F, Rossini F, Maffé P, Di Bella L. The pineal hormone melatonin in hematology and its potential efficacy in the treatment of thrombocytopenia. Recenti Prog Med. 1996; 87: 582–5.
  • Lissoni P, Tancini G, Barni S, Paolorossi F, Ardizzoia A, Conti A, Maestroni G. Treatment of cancer chemotherapy-induced toxicity with the pineal hormone melatonin. Support Care Cancer. 1997; 5: 126–9.
  • Lissoni P, Barni S, Mandalà M, Ardizzoia A, Paolorossi F, Vaghi M, Longarini R, Malugani F, Tancini G. Decreased toxicity and increased efficacy of cancer chemotherapy using the pineal hormone melatonin in metastatic solid tumour patients with poor clinical status. Eur J Cancer. 1999; 35: 1688–92.
  • Lissoni P, Mandala M, Rossini F, Fumagalli L, Barni S. Growth factors: thrombopoietic property of the pineal hormone melatonin. Hematology. 1999; 4: 335–343.
  • Lissoni P, Tancini G, Paolorossi F, Mandalà M, Ardizzoia A, Malugani F, Giani L, Barni S. Chemoneuroendocrine therapy of metastatic breast cancer with persistent thrombocytopenia with weekly low-dose epirubicin plus melatonin: a phase II study. J Pineal Res. 1999; 26: 169–73.
  • Lissoni P. Is there a role for melatonin in supportive care?. Support Care Cancer. 2002; 10: 110–6.
  • Whitnall MH, Elliott TB, Harding RA, Inal CE, Landauer MR, Wilhelmsen CL, McKinney L, Miner VL, Jackson WE3rd, Loria RM, Ledney GD, Seed TM. Androstenediol stimulates myelopoiesis and enhances resistance to infection in gamma-irradiated mice. Int J Immunopharmacol. 2000; 22: 1–14.
  • Whitnall MH, Inal CE, Jackson WE 3rd, Miner VL, Villa V, Seed TM. In vivo radioprotection by 5-androstenediol: stimulation of the innate immune system. Radiat Res. 2001; 156: 283–93.
  • Stickney DR, Dowding C, Garsd A, Ahlem C, Whitnall M, McKeon M, Reading C, Frincke J. 5-androstenediol stimulates multilineage hematopoiesis in rhesus monkeys with radiation-induced myelosuppression. Int Immunopharmacol. 2006; 6: 1706–13.
  • Stickney DR, Dowding C, Authier S, Garsd A, Onizuka-Handa N, Reading C, Frincke JM. 5-androstenediol improves survival in clinically unsupported rhesus monkeys with radiation-induced myelosuppression. Int Immunopharmacol. 2007: 500–5.
  • Gudkov SV, Gudkova OY, Chernikov AV, Bruskov VI. Protection of mice against X-ray injuries by the post-irradiation administration of guanosine and inosine. Int J Radiat Biol. 2009; 85: 116–25.
  • Gudkov SV, Gudkova OY, Chernikov AV, Bruskov VI. Tocopherol succinate: a promising radiation countermeasure. Int J Radiat Biol. 2009; 85: 116–25.
  • Akushevich IV, Veremeyeva GA, Dimov GP, Ukraintseva SV, Arbeev KG, Akleyev AV, Yashin AI. Modeling deterministic effects in hematopoietic system caused by chronic exposure to ionizing radiation in large human cohorts. Health Phys. 2010; 99: 322–9.
  • Akushevich IV, Veremeyeva GA, Dimov GP, Ukraintseva SV, Arbeev KG, Akleyev AV, Yashin AI. Modeling hematopoietic system response caused by chronic exposure to ionizing radiation. Radiat Environ Biophys. 2011; Jan 23. [Epub ahead of print].
  • Lissoni P, Bucovec R, Bonfanti A, Giani L, Mandelli A, Roselli MG, Rovelli F, Fumagalli L. Thrombopoietic properties of 5-methoxytryptamine plus melatonin versus melatonin alone in the treatment of cancer-related thrombocytopenia. J Pineal Res. 2001; 30: 123–6.
  • Lissoni P. Biochemotherapy with immunomodulating pineal hormones other than melatonin: 5-methoxytryptamine as a new oncostatic pineal agent. Pathol Biol (Paris). 2007; 55: 198–200.
  • Chahbouni M, Escames G, Venegas C, Sevilla B, García JA, López LC, Muñoz-Hoyos A, Molina-Carballo A, Acuña-Castroviejo D. Melatonin treatment normalizes plasma pro-inflammatory cytokines and nitrosative/oxidative stress in patients suffering from Duchenne muscular dystrophy. J Pineal Res. 2010; 48: 282–9.
  • Okinaga K, Iinuma H, Kitamura Y, Yokohata T, Inaba T, Fukushima R. Effect of immunotherapy and spleen preservation on immunological function in patients with gastric cancer. J Exp Clin Cancer Res. 2006; 25: 339–49.
  • Chen IJ, Yen CF, Lin KJ, Lee CL, Soong YK, Lai CH, Lin CT. Vaccination with OK-432 followed by TC-1 tumor lysate leads to significant antitumor effects. Reprod Sci. 2011. Feb 14. [Epub ahead of print].
  • Gerner EW, Tome ME, Fry SE, Bowden GT. Inhibition of ionizing radiation recovery processes in polyamine-depleted Chinese hamster cells. Cancer Res. 1988; 48: 4881–5.
  • Snyder RD. Inhibition of X-ray-induced DNA strand break repair in polyamine-depleted HeLa cells. Int J Radiat Biol. 1989; 55: 773–82.
  • Snyder RD, Lachmann PJ. Hyperthermia, polyamine depletion, and inhibition of X-ray-induced DNA strand break repair. Radiat Res. 1989; 120: 121–8.
  • Held KD, Awad S. Effects of polyamines and thiols on the radiation sensitivity of bacterial transforming DNA. Int J Radiat Biol. 1991; 59: 699–710.
  • Snyder RD, Schroeder KK. Radiosensitivity of polyamine-depleted HeLa cells and modulation by the aminothiol WR-1065. Radiat Res. 1994; 137: 67–75.
  • Williams JR, Casero RA, Dillehay LE. The effect of polyamine depletion on the cytotoxic response to PUVA, gamma rays and UVC in V79 cells in vitro. Biochem Biophys Res Commun. 1994; 201: 1–7.
  • Spotheim-Maurizot M, Ruiz S, Sabattier R, Charlier M. Radioprotection of DNA by polyamines. Int J Radiat Biol. 1995; 68: 571–7.
  • Newton GL, Aguilera JA, Ward JF, Fahey RC. Polyamine-induced compaction and aggregation of DNA-a major factor in radioprotection of chromatin under physiological conditions. Radiat Res. 1996; 145: 776–80.
  • Chiu S, Oleinick NL. Radioprotection against the formation of DNA double-strand breaks in cellular DNA but not native cellular chromatin by the polyamine spermine. Radiat Res. 1997; 148: 188–92.
  • Newton GL, Aguilera JA, Ward JF, Fahey RC. Effect of polyamine-induced compaction and aggregation of DNA on the formation of radiation-induced strand breaks: quantitative models for cellular radiation damage. Radiat Res. 1997; 148: 272–84.
  • Chiu S, Oleinick NL. Radioprotection of cellular chromatin by the polyamines spermine and putrescine: preferential action against formation of DNA-protein crosslinks. Radiat Res. 1998; 149: 543–9.
  • Ha HC, Yager JD, Woster PA, Casero RA Jr. Structural specificity of polyamines and polyamine analogues in the protection of DNA from strand breaks induced by reactive oxygen species. Biochem Biophys Res Commun. 1998; 244: 298–303.
  • Sy D, Hugot S, Savoye C, Ruiz S, Charlier M, Spotheim-Maurizot M. Radioprotection of DNA by spermine: a molecular modelling approach. Int J Radiat Biol. 1999; 75: 953–61.
  • Warters RL, Newton GL, Olive PL, Fahey RC. Radioprotection of human cell nuclear DNA by polyamines: radiosensitivity of chromatin is influenced by tightly bound spermine. Radiat Res. 1999; 151: 354–62.
  • Douki T, Bretonniere Y, Cadet J. Protection against radiation-induced degradation of DNA bases by polyamines. Radiat Res. 2000; 153: 29–35.
  • Kitada M, Igarashi K, Hirose S, Kitagawa H. Inhibition by polyamines of lipid peroxide formation in rat liver microsomes. Biochem Biophys Res Commun. 1979; 87: 388–94.
  • Kitada M, Naito Y, Igarashi K, Hirose S, Kanakubo Y, Kitagawa H. Possible mechanism of inhibition by polyamines of lipid peroxidation in rat liver microsomes. Res Commun Chem Pathol Pharmacol. 1981; 33: 487–97.
  • Ohmori S, Misaizu T, Kitada M, Kitagawa H, Igarashi K, Hirose S, Kanakubo Y. Polyamine lowered the hepatic lipid peroxide level in rats. Res Commun Chem Pathol Pharmacol. 1988; 62: 235–49.
  • Løvaas E, Carlin G. Spermine: an anti-oxidant and anti-inflammatory agent. Free Radic Biol Med. 1991; 11: 455–61.
  • Khan AU, Di Mascio P, Medeiros MH, Wilson T. Spermine and spermidine protection of plasmid DNA against single-strand breaks induced by singlet oxygen. Proc Natl Acad Sci U S A. 1992; 89: 11428–30.
  • Khan AU, Mei YH, Wilson T. A proposed function for spermine and spermidine: protection of replicating DNA against damage by singlet oxygen. Proc Natl Acad Sci U S A. 1992; 89: 11426–7.
  • Pavlovic DD, Uzunova P, Galabova T, Peneva V, Sokolova Z, Bjelakovic G, Ribarov S. Polyamines as modulators of lipoperoxidation. Gen Physiol Biophys. 1992; 11: 203–11.
  • Matkovics B, Kecskemeti V, Varga SI, Novak Z, Kertesz Z. Antioxidant properties of di- and polyamines. Comp Biochem Physiol B. 1993; 104: 475–9.
  • Løvaas E. Hypothesis: spermine may be an important epidermal antioxidant. Med Hypotheses. 1995; 45: 59–67.
  • Løvaas E. Antioxidative and metal-chelating effects of polyamines. Adv Pharmacol. 1997; 38: 119–49.
  • Ha HC, Sirisoma NS, Kuppusamy P, Zweier JL, Woster PM, Casero RA Jr. The natural polyamine spermine functions directly as a free radical scavenger. Proc Natl Acad Sci U S A. 1998; 95: 11140–5.
  • Grudziński IP, Frankiewicz-Józko A. Further studies on the anti-oxidative effect of putrescine in sodium nitrite-treated rats. Rocz Panstw Zakl Hig. 2002; 53: 11–7.
  • Farriol M, Segovia-Silvestre T, Venereo Y, Orta X. Antioxidant effect of polyamines on erythrocyte cell membrane lipoperoxidation after free-radical damage. Phytother Res. 2003; 17: 44–7.
  • Bellé NA, Dalmolin GD, Fonini G, Rubin MA, Rocha JB. Polyamines reduces lipid peroxidation induced by different pro-oxidant agents. Brain Res. 2004; 1008: 245–51.
  • Das KC, Misra HP. Hydroxyl radical scavenging and singlet oxygen quenching properties of polyamines. Mol Cell Biochem. 2004; 262: 127–33.
  • Fujisawa S, Kadoma Y. Kinetic evaluation of polyamines as radical scavengers. Anticancer Res. 2005; 25: 965–9.
  • von Deutsch AW, Mitchell CD, Williams CE, Dutt K, Silvestrov NA, Klement BJ, Abukhalaf IK, von Deutsch DA. Polyamines protect against radiation-induced oxidative stress. Gravit Space Biol Bull. 2005; 18: 109–10.
  • Hernández SM, Sánchez MS, de Tarlovsky MN. Polyamines as a defense mechanism against lipoperoxidation in Trypanosoma cruzi. Acta Trop. 2006; 98: 94–102.
  • Sava IG, Battaglia V, Rossi CA, Salvi M, Toninello A. Free radical scavenging action of the natural polyamine spermine in rat liver mitochondria. Free Radic Biol Med. 2006; 41: 1272–81.
  • Rider JE, Hacker A, Mackintosh CA, Pegg AE, Woster PM, Casero RAJr. Spermine and spermidine mediate protection against oxidative damage caused by hydrogen peroxide. Amino Acids. 2007; 33: 231–40.
  • Mozdzan M, Szemraj J, Rysz J, Stolarek RA, Nowak D. Anti-oxidant activity of spermine and spermidine re-evaluated with oxidizing systems involving iron and copper ions. Int J Biochem Cell Biol. 2006; 38: 69–81.
  • Bardócz S, Duguid TJ, Brown DS, Grant G, Pusztai A, White A, Ralph A. The importance of dietary polyamines in cell regeneration and growth. Br J Nutr. 1995; 73: 819–28.
  • Wallace HM. The polyamines: past, present and future. Essays Biochem. 2009; 46: 1–9.
  • Landau G, Bercovich Z, Park MH, Kahana C. The role of polyamines in supporting growth of mammalian cells is mediated through their requirement for translation initiation and elongation. J Biol Chem. 2010; 285: 12474–81.
  • Pollack PF, Koldovsk? O, Nishioka K. Polyamines in human and rat milk and in infant formulas. Am J Clin Nutr. 1992; 56: 371–5.
  • Romain N, Dandrifosse G, Jeusette F, Forget P. Polyamine concentration in rat milk and food, human milk, and infant formulas. Pediatr Res. 1992; 32: 58–63.
  • Buts JP, De Keyser N, De Raedemaeker L, Collette E, Sokal EM. Polyamine profiles in human milk, infant artificial formulas, and semi-elemental diets. J Pediatr Gastroenterol Nutr. 1995; 21: 44–9.
  • Dorhout B, van Beusekom CM, Huisman M, Kingma AW, de Hoog E, Boersma ER, Muskiet FA. Estimation of 24-hour polyamine intake from mature human milk. J Pediatr Gastroenterol Nutr. 1996; 23: 298–302.
  • Löser C. Polyamines in human and animal milk. Br J Nutr. 2000; 1(84 Suppl): S55–8.
  • Larqué E, Sabater-Molina M, Zamora S. Biological significance of dietary polyamines. Nutrition. 2007; 23: 87–95.
  • Wang JY, McCormack SA, Viar MJ, Johnson LR. Stimulation of proximal small intestinal mucosal growth by luminal polyamines. Am J Physiol. 1991; 261: G504–11.
  • Buts JP, De Keyser N, Kolanowski J, Sokal E, Van Hoof F. Maturation of villus and crypt cell functions in rat small intestine. Role of dietary polyamines. Dig Dis Sci. 1993; 38: 1091–8.
  • Wéry I, Dandrifosse G. Evolution of biochemical parameters characterizing the proximal small intestine after orally administered spermine in unweaned rats. Endocr Regul. 1993; 27: 201–7.
  • Harada E, Hashimoto Y, Syuto B. Orally administered spermine induces precocious intestinal maturation of macromolecular transport and disaccharidase development in suckling rats. Comp Biochem Physiol A Physiol. 1994; 109: 667–73.
  • Kaouass M, Deloyer P, Dandrifosse G. Intestinal development in suckling rats: direct or indirect spermine action?. Digestion. 1994; 55: 160–7.
  • Kaouass M, Deloyer P, Wery I, Dandrifosse G. Analysis of structural and biochemical events occurring in the small intestine after dietary polyamine ingestion in suckling rats. Dig Dis Sci. 1996; 41: 1434–44.
  • Wéry I, Deloyer P, Dandrifosse G. Effects of a single dose of orally-administered spermine on the intestinal development of unweaned rats. Arch Physiol Biochem. 1996; 104: 163–72.
  • Kaouass M, Deloyer P, Gouders I, Peulen O, Dandrifosse G. Role of interleukin-1 beta, interleukin-6, and TNF-alpha in intestinal maturation induced by dietary spermine in rats. Endocrine. 1997; 6: 187–94.
  • Capano G, Bloch KJ, Carter EA, Dascoli JA, Schoenfeld D, Harmatz PR. Polyamines in human and rat milk influence intestinal cell growth in vitro. J Pediatr Gastroenterol Nutr. 1998; 27: 281–6.
  • Patel AR, Li J, Bass BL, Wang JY. Expression of the transforming growth factor-beta gene during growth inhibition following polyamine depletion. Am J Physiol. 1998; 275: C590–8.
  • Rao JN, Li J, Li L, Bass BL, Wang JY. Differentiated intestinal epithelial cells exhibit increased migration through polyamines and myosin II. Am J Physiol. 1999; 277: G1149–58.
  • Greco S, Hugueny I, George P, Perrin P, Louisot P, Biol MC. Influence of spermine on intestinal maturation of the glycoprotein glycosylation process in neonatal rats. Biochem J. 2000; 345: 69–75.
  • Peulen O, Dandrifosse G. Cyclosporine A inhibits partially spermine-induced differentiation but not cell loss of suckling rat small intestine. Dig Dis Sci. 2000; 45: 750–4.
  • Gréco S, Niepceron E, Hugueny I, George P, Louisot P, Biol MC. Dietary spermidine and spermine participate in the maturation of galactosyltransferase activity and glycoprotein galactosylation in rat small intestine. J Nutr. 2001; 131: 1890–7.
  • Rao JN, Li L, Golovina VA, Platoshyn O, Strauch ED, Yuan JX, Wang JY. Ca2 + -RhoA signaling pathway required for polyamine-dependent intestinal epithelial cell migration. Am J Physiol Cell Physiol. 2001; 280: C993–1007.
  • Biol-N'Garagba MC, Greco S, George P, Hugueny I, Louisot P. Polyamine participation in the maturation of glycoprotein fucosylation, but not sialylation, in rat small intestine. Pediatr Res. 2002; 51: 625–34.
  • Rao JN, Guo X, Liu L, Zou T, Murthy KS, Yuan JX, Wang JY. Polyamines regulate Rho-kinase and myosin phosphorylation during intestinal epithelial restitution. Am J Physiol Cell Physiol. 2003; 284: C848–59.
  • Peulen O, Dandrifosse G. Spermine-induced maturation in Wistar rat intestine: a cytokine-dependent mechanism. J Pediatr Gastroenterol Nutr. 2004; 38: 524–32.
  • Rao JN, Liu L, Zou T, Marasa BS, Boneva D, Wang SR, Malone DL, Turner DJ, Wang JY. Polyamines are required for phospholipase C-gamma1 expression promoting intestinal epithelial restitution after wounding. Am J Physiol Gastrointest Liver Physiol. 2007; 292: G335–43.
  • Sabater-Molina M, Larqué E, Torrella F, Plaza J, Lozano T, Muñoz A, Zamora S. Effects of dietary polyamines at physiologic doses in early-weaned piglets. Nutrition. 2009; 25: 940–6.
  • Leroy D, Schmid N, Behr JP, Filhol O, Pares S, Garin J, Bourgarit JJ, Chambaz EM, Cochet C. Direct identification of a polyamine binding domain on the regulatory subunit of the protein kinase casein kinase 2 by photoaffinity labeling. J Biol Chem. 1995; 270: 17400–6.
  • Leroy D, Heriché JK, Filhol O, Chambaz EM, Cochet C. Binding of polyamines to an autonomous domain of the regulatory subunit of protein kinase CK2 induces a conformational change in the holoenzyme. A proposed role for the kinase stimulation. J Biol Chem. 1997; 272: 20820–7.
  • Trembley JH, Chen Z, Unger G, Slaton J, Kren BT, Van Waes C, Ahmed K. Emergence of protein kinase CK2 as a key target in cancer therapy. Biofactors. 2010; 36: 187–95.
  • Li D, Dobrowolska G, Krebs EG. The physical association of casein kinase 2 with nucleolin. J Biol Chem. 1996; 271: 15662–8.
  • Li D, Dobrowolska G, Krebs EG. Identification of proteins that associate with protein kinase CK2. Mol Cell Biochem. 1999; 191: 223–8.
  • Li D, Meier UT, Dobrowolska G, Krebs EG. Specific interaction between casein kinase 2 and the nucleolar protein Nopp140. J Biol Chem. 1997; 272: 3773–9.
  • Szebeni A, Hingorani K, Negi S, Olson MO. Role of protein kinase CK2 phosphorylation in the molecular chaperone activity of nucleolar protein B23. J Biol Chem. 2003; 278: 9107–15.
  • Bandyopadhyay K, Gjerset RA. Protein kinase CK2 is a central regulator of topoisomerase I hyperphosphorylation and camptothecin sensitivity in cancer cell lines. Biochemistry. 2011; 50: 704–14.
  • Paytubi S, Morrice NA, Boudeau J, Proud CG. The N-terminal region of ABC50 interacts with eukaryotic initiation factor eIF2 and is a target for regulatory phosphorylation by CK2. Biochem J. 2008; 409: 223–31.
  • Ampofo E, Kietzmann T, Zimmer A, Jakupovic M, Montenarh M, Götz C. Phosphorylation of the von Hippel-Lindau protein (VHL) by protein kinase CK2 reduces its protein stability and affects p53 and HIF-1alpha mediated transcription. Int J Biochem Cell Biol. 2010; 42: 1729–35.
  • Laitinen PH, Hietala OA, Pulkka AE, Pajunen AE. Purification of mouse brain ornithine decarboxylase reveals its presence as an inactive complex with antizyme. Biochem J. 1986; 236: 613–6.
  • Kilpeläinen PT, Hietala OA. Activation of rat brain ornithine decarboxylase by GTP. Biochem J. 1994; 300: 577–82.
  • Anagnostopoulos CG, Kyriakidis DA. Regulation of the Escherichia coli biosynthetic ornithine decarboxylase activity by phosphorylation and nucleotides. Biochim Biophys Acta. 1996; 1297: 228–34.
  • Moxnes JF, Christophersen OA. Counter attacking pandemic H5N1 bird influenza by counter pandemic. Microb Ecol Health Dis. 2006; 20: 1–26.