1,327
Views
180
CrossRef citations to date
0
Altmetric
Reviews

Role Of Lipids In Brain Injury And Diseases

&
Pages 403-422 | Published online: 18 Jan 2017

Bibliography

  • Wenk MR: The emerging field of lipidomics. Nat. Rev. Drug Discov. 4(7), 594–610 (2005).
  • Excellent comprehensive review on various aspects of lipidomics and bioinformatics.
  • Fahy E, Subramaniam S, Brown HA et al.: A comprehensive classification system for lipids. J. Lipid Res. 46(5), 839–862 (2005).
  • Comprehensive classification of lipids with a common platform that is compatible with informatics requirements for the hard-core lipid community.
  • Adibhatla RM, Hatcher JF, Dempsey RJ: Lipids and lipidomics in brain injury and diseases. AAPS J. 8(2), E314–E321 (2006).
  • Adibhatla RM, Hatcher JF: Phospholipase A2, reactive oxygen species, and lipid peroxidation in cerebral ischemia. Free Radic. Biol. Med. 40(3), 376–387 (2006).
  • Jiang J, Borisenko GG, Osipov A et al.: Arachidonic acid-induced carbon-centered radicals and phospholipid peroxidation in cyclo-oxygenase-2-transfected PC12 cells. J. Neurochem. 90(5), 1036–1049 (2004).
  • Simmons DL, Botting RM, Hla T: Cyclooxygenase isozymes: The biology of prostaglandin synthesis and inhibition. Pharmacol Rev. 56(3), 387–437 (2004).
  • Comprehensive review on cyclooxygenease (COX) enzymes and prostaglandin synthesis.
  • Kunz A, Anrather J, Zhou P, Orio M, Iadecola C: Cyclooxygenase-2 does not contribute to postischemic production of reactive oxygen species. J. Cereb. Blood Flow Metab. 27(3), 545–551 (2007).
  • First report showing that COX-2 does not contribute to reactive oxygen species (ROS) production.
  • Delacourte A, Sergeant N, Champain D et al.: Nonoverlapping but synergetic tau and APP pathologies in sporadic Alzheimer’s disease. Neurology. 59(3), 398–407 (2002).
  • Highlights the missing link between synergetic tau and A pathologies.
  • Ehehalt R, Keller P, Haass C, Thiele C, Simons K: Amyloidogenic processing of the Alzheimer -amyloid precursor protein depends on lipid rafts. J. Cell Biol. 160(1), 113–123 (2003).
  • Suggests that cleavage of the different amyloid precursor protein (APP) pools by α- versus β-secretases depends on distribution of APP to lipid rafts.
  • Mandavilli A: The amyloid code. Nat. Med. 12(7), 747–751 (2006).
  • Provocative commentary on the current amyloid theories in Alzheimer’s disease (AD).
  • Herrup K, Neve R, Ackerman SL, Copani A: Divide and die: Cell cycle events as triggers of nerve cell death. J. Neurosci. 24(42), 9232–9239 (2004).
  • Critical review on neuronal cell cycle and death and transgenic models in AD.
  • Schwab C, Hosokawa M, McGeer PL: Transgenic mice over-expressing amyloid beta protein are an incomplete model of Alzheimer disease. Exp. Neurol. 188(1), 52–64 (2004).
  • Transgenic models over-expressing Aβ alone are incomplete models for AD and have limited use.
  • Gotz J, Streffer JR, David D et al.: Transgenic animal models of Alzheimer’s disease and related disorders: histopathology, behavior and therapy. Mol. Psychiatry 9(7), 664–683 (2004).
  • Puglielli L: Aging of the brain, neurotrophin signaling, and Alzheimer’s disease: Is IGF1- R the common culprit? Neurobiol. Aging doi: 10.1016/j.neurobiolaging.2007.01.010 (2007).
  • Puglielli L, Tanzi RE, Kovacs DM: Alzheimer’s disease: the cholesterol connection. Nature Neurosci. 6(4), 345–351 (2003).
  • Presents the connection between cholesterol and Aβ and how statins lower Aβ in experimental models by inhibiting cholesterol synthesis.
  • Whitfield JF: Can statins put the brakes on Alzheimer’s disease? Expert Opin. Investig. Drugs. 15(12), 1479–1485 (2006).
  • Review suggests more clinical trials are needed to specifically assess the statin effects on sporadic AD.
  • Grimm MOW, Grimm HS, Patzold AJ et al.: Regulation of cholesterol and sphingomyelin metabolism by amyloid- and presenilin. Nat. Cell Biol. 7(11), 1118–1123 (2005).
  • Model suggesting the combined effects of cellular cholesterol and sphingomyelin levels regulate α-secretase activity in APP cleavage.
  • Mattson MP, Cutler RG, Jo DG: Alzheimer peptides perturb lipid-regulating enzymes. Nat. Cell Biol. 7(11), 1045–1047 (2005).
  • Caballero J, Nahata M: Do statins slow down Alzheimer’s disease? A review. J. Clin. Pharm. Ther. 29(3), 209–213 (2004).
  • Moses GS, Jensen MD, Lue LF et al.: Secretory PLA2-IIA: A new inflammatory factor for Alzheimer’s disease. J. Neuroinflammation. 3(1), doi: 10.1186/1742–2094–1183–1128 (2006).
  • First report showing the expression of sPLA2 IIA (inflammatory PLA2) in AD brains.
  • Williams TI, Lynn BC, Markesbery WR, Lovell MA: Increased levels of 4- hydroxynonenal and acrolein, neurotoxic markers of lipid peroxidation, in the brain in mild cognitive impairment and early Alzheimer’s disease. Neurobiol. Aging 27(8), 1094–1099 (2006).
  • Liu X, Lovell MA, Lynn BC: Development of a method for quantification of acroleindeoxyguanosine adducts in DNA using isotope dilution-capillary LC/MS/MS and its application to human brain tissue. Anal. Chem. 77(18), 5982–5989 (2005).
  • Mariani E, Polidori MC, Cherubini A, Mecocci P: Oxidative stress in brain aging, neurodegenerative and vascular diseases: an overview. J. Chromatog. B. 827(1), 65–75 (2005).
  • Oddo S, Billings L, Kesslak JP, Cribbs DH, LaFerla FM: A immunotherapy leads to clearance of early, but not late, hyperphosphorylated tau aggregates via the proteasome. Neuron 43(3), 321–332 (2004).
  • Farooqui AA, Ong W-Y, Horrocks LA: Inhibitors of brain phospholipase A2 activity: their neuropharmacological effects and therapeutic importance for the treatment of neurologic disorders. Pharmacol Rev. 58(3), 591–620 (2006).
  • Hauser RA, Zesiewicz TA: Advances in the pharmacologic management of early Parkinson disease. Neurologist 13(3), 126–132 (2007).
  • Welch K, Yuan J: -Synuclein oligomerization: a role for lipids? Trends Neurosci. 26(10), 517–519 (2003).
  • Discusses the relationship between polyunsaturated fatty acid (PUFA) and α-synuclein multimerization in PD.
  • Sharon R, Bar-Joseph I, Frosch MP, Walsh DM, Hamilton JA, Selkoe DJ: The formation of highly soluble oligomers of - synuclein is regulated by fatty acids and enhanced in Parkinson’s disease. Neuron. 37(4), 583–595 (2003).
  • Samadi P, Grègoire L, Rouillard C et al.: Docosahexaenoic acid reduces levodopainduced dyskinesias in 1-methyl-4-phenyl- 1,2,3,6-tetrahydropyridine monkeys. Ann. Neurol. 59(2), 282–288 (2006).
  • Horinouchi K, Erlich S, Perl DP et al.: Acid sphingomyelinase deficient mice - a model of types A and B Niemann-Pick disease. Nature Genetics 10(3), 288–293 (1995).
  • An animal model for Niemann-Pick types A and B diseases was developed.
  • Futerman AH, van Meer G: The cell biology of lysosomal storage disorders. Nat. Rev. Mol. Cell Biol. 5(7), 554–565 (2004).
  • Larsen EC, Hatcher JF, Adibhatla RM: Effect of tricyclodecan-9-yl potassium xanthate (D609) on phospholipid metabolism and cell death during oxygenglucose deprivation in PC12 cells. Neuroscience 146(3), 946–961 (2007).
  • Report showing that the loss of phosphatidylcholine (PC) is a cause rather than a result of cell death.
  • Vance JE: Lipid imbalance in the neurological disorder, Niemann-Pick C disease. FEBS Lett. 580(23), 5518–5524 (2006).
  • Maxfield FR, Tabas I: Role of cholesterol and lipid organization in disease. Nature 438(7068), 612–621 (2005).
  • This review is more biophysical in nature and discusses the importance of cholesterol in membrane phospholipid organization.
  • Aarli JA: Role of cytokines in neurological disorders. Curr. Med. Chem. 10, 1931–1937 (2003).
  • Carlson NG, Rose JW: Antioxidants in multiple sclerosis: do they have a role in therapy? CNS Drugs 20(6), 433–441 (2006).
  • Kalyvas A, David S: Cytosolic phospholipase A2 plays a key role in the pathogenesis of multiple sclerosis-like disease. Neuron. 41(3), 323–335 (2004).
  • First report showing the cytosolic phospholipase A2 (cPLA2) role in multiple sclerosis (MS).
  • Marusic S, Leach MW, Pelker JW et al.: Cytosolic phospholipase A2a-deficient mice are resistant to experimental autoimmune encephalomyelitis. J. Exp. Med. 202(6), 841–851 (2005).
  • Heller RA, Kronke M: TNF receptormediated signaling pathways. J. Cell Biol. 126, 5–9 (1994).
  • Kronke M, Adam-Klages S: Role of caspases in TNF-mediated regulation of cPLA2. FEBS Lett. 531(1), 18–22 (2002).
  • Cunningham TJ, Yao L, Oetinger M, Cort L, Blankenhorn EP, Greensteinm JI: Secreted phospholipase A2 activity in experimental autoimmune encephalomyelitis and multiple sclerosis. J. Neuroinflammation 3 doi: 10.1186/1742–2094–3-26, (2006).
  • Aktas O, Waiczies S, Zipp F: Neurodegeneration in autoimmune demyelination: Recent mechanistic insights reveal novel therapeutic targets. J. Neuroimmunol. 184(1–2), 17–26 (2007).
  • Maccarrone M, Battista N, Centonze D: The endocannabinoid pathway in Huntington’s disease: A comparison with other neurodegenerative diseases. Prog. Neurobiol. 81(5–6), 349–379 (2007).
  • Review discusses the endocannabinoid pathway in AD, parkinson’s disease (PD), Huntington disease (HD) and MS. Agonists/antagonists of endocannabinoid receptors and inhibitors of endocannabinoid metabolism are discussed in detail.
  • Pacher P, Batkai S, Kunos G: The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacol. Rev. 58(3), 389–462 (2006).
  • Degroot A, Nomikos GG: In vivo neurochemical effects induced by changes in endocannabinoid neurotransmission. Curr. Opin. Pharmacol. 7(1), 62–68 (2007).
  • Battista N, Fezza F, Finazzi-Agro A, Maccarrone M: The endocannabinoid system in neurodegeneration. Ital. J. Biochem. 55(3–4), 283–289 (2006).
  • Clifford JJ, Drago J, Natoli AL et al.: Essential fatty acids given from conception prevent topographies of motor deficit in a transgenic model of Huntington’s disease. Neuroscience 109(1), 81–88 (2002).
  • Puri BK, Leavitt BR, Hayden MR et al.: Ethyl-EPA in Huntington disease: a double-blind, randomized, placebocontrolled trial. Neurology 65(2), 286–292 (2005).
  • Muma NA: Transglutaminase is linked to neurodegenerative diseases. J. Neuropathol. Exp. Neurol. 66(4), 258–263 (2007).
  • Stack EC, Smith KM, Ryu H et al.: Combination therapy using minocycline and coenzyme Q10 in R6/2 transgenic Huntington’s disease mice. Biochim. Biophys. Acta. 1762(3), 373–380 (2006).
  • Kunst CB: Complex genetics of amyotrophic lateral sclerosis. Am. J. Hum. Genet. 75(6), 933–947 (2004).
  • Thorough review of amyotrophic lateral sclerosis (ALS) genetics.
  • Bruijn LI, Miller TM, Cleveland DW: Unraveling the mechanisms involved in motor neuron degeneration in ALS. Ann. Rev. Neurosci. 27(1), 723–749 (2004).
  • Nirmalananthan N, Greensmith L: Amyotrophic lateral sclerosis: recent advances and future therapies. Curr. Opin. Neurol. 18(6), 712–719 (2005).
  • This articles discusses clinical trials in ALS. Despite intense research, riluzole, an antiglutamate agent, still remains the only treatment for ALS.
  • Minghetti L: Cyclooxygenase-2 (COX-2) in inflammatory and degenerative brain diseases. J. Neuropath. Exp. Neurol. 63(9), 901–910 (2004).
  • Agar J, Durham H: Relevance of oxidative injury in the pathogenesis of motor neuron diseases. Amyotroph. Lateral Scler. Other Motor Neuron Disord. 4(4), 232–242 (2003).
  • Simpson EP, Henry YK, Henkel JS, Smith RG, Appel SH: Increased lipid peroxidation in sera of ALS patients: A potential biomarker of disease burden. Neurology 62(10), 1758–1765 (2004).
  • Horrobin D: The lipid hypothesis of schizophrenia. In: Brain Lipids and Disorders in Biological Psychiatry. Skinner ER (Ed.), Elsevier Science, Amsterdam, The Netherlands 39–52 (2002).
  • Role of lipids in schizophrenia is elegantly presented in this book chapter.
  • Berger GE, Smesny S, Amminger GP: Bioactive lipids in schizophrenia. Int. Rev. Psychiatry. 18(2), 85–98 (2006).
  • Hakak Y, Walker JR, Li C et al.: Genome-wide expression analysis reveals dysregulation of myelination-related genes in chronic schizophrenia. Proc. Natl. Acad. Sci. USA 98(8), 4746–4751 (2001).
  • Papandreou D, Pavlou E, Kalimeri E, Mavromichalis I: The ketogenic diet in children with epilepsy. Br. J. Nutr. 95(1), 5–13 (2006).
  • LaRoche SM: A new look at the secondgeneration antiepileptic drugs: a decade of experience. Neurologist. 13(3), 133–139 (2007).
  • Bialer M, Johannessen SI, Kupferberg HJ, Levy RH, Perucca E, Tomson T: Progress report on new antiepileptic drugs: a summary of the Eigth Eilat Conference (EILAT VIII). Epilepsy Research 73(1), 1–52 (2007).
  • Review of the new generation anti-epileptic drugs.
  • Klotz U: The role of pharmacogenetics in the metabolism of antiepileptic drugs: pharmacokinetic and therapeutic implications. Clin. Pharmacokinet. 46(4), 271–279 (2007).
  • Gasior M, Rogawski MA, Hartman AL: Neuroprotective and disease-modifying effects of the ketogenic diet. Behav. Pharmacol. 17(5–6), 431–439 (2006).
  • Bough KJ, Rho JM: Anticonvulsant mechanisms of the ketogenic diet. Epilepsia. 48(1), 43–58 (2007).
  • Presents a view of how the ketogenic diet exerts anticonvulsant effects, summarizing key insights from experimental and clinical studies. PUFAs are generated after ketogenic diet that up-regulate various energy metabolism genes and mitochondrial biogenesis that reduces ROS generation and increase energy production.
  • Savitz SI: A critical appraisal of the NXY- 059 neuroprotection studies for acute stroke: A need for more rigorous testing of neuroprotective agents in animal models of stroke. Exp. Neurol. 205(1), 20–25 (2007).
  • A critical appraisal of failed Stroke Acute Ischemic NXY-059 Treatment (SAINT) II Phase III stroke clinical trial.
  • Adibhatla RM, Hatcher JF: Cytidine 5’- diphosphocholine (CDP-choline) in stroke and other CNS disorders. Neurochem. Res. 30(1), 15–23 (2005).
  • Review on CDP-choline actions in stroke and its possible effect in regulating proinflammatory cytokines such as TNF-α and IL-1β.
  • Adibhatla RM, Hatcher JF, Larsen EC, Chen X, Sun D, Tsao F: CDP-choline significantly restores the phosphatidylcholine levels by differentially affecting phospholipase A2 and CTPphosphocholine cytidylyltransferase after stroke. J. Biol. Chem. 281(10), 6718–6725 (2006).
  • First report showing the loss of PC due to activation of sPLA2 and loss of CCT; CDPcholine restored PC levels by opposing these actions in stroke model.
  • Mehta SL, Manhas N, Raghubir R: Molecular targets in cerebral ischemia for developing novel therapeutics. Brain Res. Rev. 54(1) 34–66, (2007).
  • Comprehensive review of molecular mechanisms in cerebral ischemia.
  • Liu T, Clark RK, McDonnell PC et al.: TNF- expression in ischemic neurons. Stroke 25(7), 1481–1488 (1994).
  • Wang CX, Shuaib A: Involvement of inflammatory cytokines in central nervous system injury. Prog. Neurobiol. 67(2), 161–172 (2002).
  • Loddick SA, Rothwell NJ: Neuroprotective effects of human recombinant interleukin-1 receptor antagonist in focal cerebral ischaemia in the rat. J. Cereb. Blood Flow Metab. 16(5), 932–940 (1996).
  • Wang X, Yue TL, Barone FC, White RF, Gagnon RC, Feuerstein GZ: Concomitant cortical expression of TNF- and IL-1 mRNAs follows early response gene expression in transient focal ischemia. Mol. Chem. Neuropath. 23(2–3), 103–114 (1994).
  • Adibhatla RM, Dempsey RJ, Hatcher JF: Integration of cytokine biology and lipid metabolism in stroke. Front Neurosurg. Res. (under the aegis of Front Biosci). (2007) (In Press).
  • Secades JJ, Lorenzo JL: Citicoline: Pharmacological and clinical review, 2006 update. Methods Find. Exp. Clin. Pharmacol. 28, 1–56 (2006).
  • Savitz SI, Fisher M: Future of neuroprotection for acute stroke: In the aftermath of the SAINT trials. Ann. Neurol. 61(5), 396–402 (2007).
  • Critical issues and Stroke Therapy Academic Industrial Roundtable (STAIR) guidelines for future stroke clinical trials are discussed in the aftermath of failed SAINT-II NXY-059 stroke trails.
  • Crack PJ, Taylor JM: Reactive oxygen species and the modulation of stroke. Free Radic. Biol. Med. 38(11), 1433–1444 (2005).
  • Liu KJ, Rosenberg GA: Matrix metalloproteinases and free radicals in cerebral ischemia. Free Radic. Biol. Med. 39(1), 71–80 (2005).
  • Margaill I, Plotkine M, Lerouet D: Antioxidant strategies in the treatment of stroke. Free Radic. Biol. Med. 39(4), 429–443 (2005).
  • Tomitori H, Usui T, Saeki N et al.: Polyamine oxidase and acrolein as novel biochemical markers for diagnosis of cerebral stroke. Stroke 36(12), 2609–2613 (2005).
  • First report showing acrolein, a highly toxic aldehyde (100-times more potent than HNE), in plasma of stroke patients. So far accumulation of acrolein in stroke brains had not been demonstrated.
  • Rigg JL, Zafonte RD: Corticosteroids in TBI: is the story closed? J. Head Trauma Rehab. 21(3), 285–288 (2006).
  • Ariza M, Pueyo R, Matarin MdM et al.: Influence of APOE polymorphism on cognitive and behavioral outcome in moderate and severe traumatic brain injury. J. Neurol. Neurosurg. Psychiatry. 77(10), 1191–1193 (2006).
  • Interesting clinical study demonstrating that performance of neuropsychological tasks is worse in patients with APOE 4 allele after TBI.
  • Iwata A, Browne KD, Chen X-H, Yuguchi T, Smith DH: Traumatic brain injury induces biphasic upregulation of ApoE and ApoJ protein in rats. J. Neurosci. Res. 82(1), 103–114 (2005).
  • Fluid percussion injury showed Aaccumulation and increased ApoE mRNA expression in rats.
  • Jellinger KA: Head injury and dementia. Curr Opin Neurol. 17(6), 719–723 (2004).
  • Starkstein SE, Jorge R: Dementia after traumatic brain injury. Int. Psychogeriatr. 17(Suppl. 1), S93–S107 (2005).
  • Smith C, Graham DI, Murray LS, Stewart J, Nicoll JAR: Association of APOE e4 and cerebrovascular pathology in traumatic brain injury. J. Neurol. Neurosurg. Psychiatry 77(3), 363–366 (2006).
  • Mahmood A, Lu D, Qu C, Goussev A, Chopp M: Treatment of traumatic brain injury with a combination therapy of marrow stromal cells and atorvastatin in rats. Neurosurgery 60(3), 546–554 (2007).
  • Lynch JR, Wang H, Mace B et al.: A novel therapeutic derived from apolipoprotein E reduces brain inflammation and improves outcome after closed head injury. Exp. Neurol. 192(1), 109–116 (2005).
  • Hall ED, Springer JE: Neuroprotection and acute spinal cord injury: a reappraisal. NeuroRx 1(1), 80–100 (2004).
  • Kim H-Y: Novel metabolism of docosahexaenoic acid in neural cells. J. Biol. Chem. 282(26), 18861–18665 (2007).
  • Marcheselli VL, Hong S, Lukiw WJ et al.: Novel docosanoids inhibit brain ischemiareperfusion- mediated leukocyte infiltration and pro-inflammatory gene expression. J. Biol. Chem. 278(44), 43807–43817 (2003).
  • Bazan N: The onset of brain injury and neurodegeneration triggers the synthesis of docosanoid neuroprotective signaling. Cell. Mol. Neurobiol. 26(4–6), 901–913 (2006).
  • Ariel A, Serhan CN: Resolvins and protectins in the termination program of acute inflammation. Trends Immunol. 28(4), 176–183 (2007).
  • Belayev L, Marcheselli VL, Khoutorova L et al.: Docosahexaenoic acid complexed to albumin elicits high-grade ischemic neuroprotection. Stroke 36(1), 118–123 (2005).
  • Lukiw WJ, Cui J-G, Marcheselli VL et al.: A role for docosahexaenoic acid-derived neuroprotectin D1 in neural cell survival and Alzheimer disease. J. Clin. Invest. 115(10), 2774–2783 (2005).
  • Balazy M: Eicosanomics: targeted lipidomics of eicosanoids in biological systems. Prostaglandins Other Lipid Mediat. 73(3–4), 173–180 (2004).
  • Hillard CJ: Lipids and drugs of abuse. Life Sci. 77(14), 1531–1542 (2005).
  • Senn C, Hangartner C, Moes S, Guerini D, Hofbauer KG: Central administration of small interfering RNAs in rats: a comparison with antisense oligonucleotides. Eur. J. Pharmacol. 522(1–3), 30–37 (2005).
  • Cejka D, Losert D, Wacheck V: Short interfering RNA (siRNA): tool or therapeutic? Clin. Sci. (Lond). 110(1), 47–58 (2006).
  • This review and the following one by Dykxhoorn et al. discuss the technology of RNA silencing, potential obstacles and possibilities for clinical applications.
  • Dykxhoorn DM, Palliser D, Lieberman J: The silent treatment: siRNAs as small molecule drugs. Gene Ther. 13, 541–552 (2006).
  • Adibhatla RM, Hatcher JF, Tureyen K: CDP-choline liposomes provide significant reduction in infarction over free CDPcholine in stroke. Brain Res. 1058(1–2), 193–197 (2005).
  • Efficient delivery of CDP-choline to the brain using liposome encapsulation substantially improves the outcome after stroke. The route of administration is very important in clinical trials, which has been ignored in US trials.
  • Adibhatla RM: Mechanistic aspects of CDP-choline (Citicoline) action in stroke. In: 4th Asia Pacific Conference Against Stroke (APCAS March 30-April 1, 2007). Conference Secretariat, APCAS New Delhi, India (2007).
  • Adibhatla RM, Hatcher JF: Citicoline mechanisms and clinical efficacy in cerebral ischemia. J. Neurosci. Res. 70(2), 133–139 (2002).
  • Wolozin B: Cholesterol, statins and dementia. Curr. Opin. Lipidol. 15(6), 667–672 (2004).
  • Lane RM, Farlow MR: Lipid homeostasis and apolipoprotein E in the development and progression of Alzheimer’s disease. J. Lipid Res. 46(5), 949–968 (2005).
  • Farooqui AA, Horrocks LA, Farooqui T: Modulation of inflammation in brain: a matter of fat. J. Neurochem. 101(3), 577–599 (2007).
  • Mukherjee S, Maxfield FR: Lipid and cholesterol trafficking in NPC. Biochim. Biophys. Acta. 1685(1–3), 28–37 (2004).
  • Miranda SRP, He X, Simonaro CM et al.: Infusion of recombinant human acid sphingomyelinase into Niemann-Pick disease mice leads to visceral, but not neurological, correction of the pathophysiology. FASEB J. 14(13), 1988–1995 (2000).
  • Kanter JL, Narayana S, Ho PP et al.: Lipid microarrays identify key mediators of autoimmune brain inflammation. Nat. Med. 12(1), 138–143 (2006).
  • Centonze D, Finazzi-Agro A, Bernardi G, Maccarrone M: The endocannabinoid system in targeting inflammatory neurodegenerative diseases. Trends Pharmacol. Sci. 28(4), 180–187 (2007).
  • Lin T-N, Wang Q, Simonyi A et al.: Induction of secretory phospholipase A2 in reactive astrocytes in response to transient focal cerebral ischemia in the rat brain. J. Neurochem. 90(3), 637–645 (2004).
  • Reid RC: Inhibitors of secretory phospholipase A2 group IIA. Curr. Med. Chem. 12(25), 3011–3026 (2005).
  • Presents the structurally diverse array of available sPLA2 group IIA inhibitors, their associated biological activity in animal models and evaluation of therapeutic potential in Phase II clinical trials.
  • Narayan RK, Michel ME, Ansell B et al.: Clinical trials in head injury. J. Neurotrauma 19(5), 503–557 (2002).
  • Despite promising preclinical data, most of the clinical TBI trials that have been performed in recent years have failed to demonstrate any significant improvement in outcomes. This report summarizes the key points of a workshop sponsored by NIH/National Institute of Neurological Disorders and Stroke (NINDS) to evaluate problems of past trials and insights into proper planning of future clinical trials.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.