2,139
Views
14
CrossRef citations to date
0
Altmetric
Reviews; Medical Biotechnology

Deregulation of the circadian clock constitutes a significant factor in tumorigenesis: a clockwork cancer. Part II. In vivo studies

&
Pages 379-386 | Received 20 Mar 2014, Accepted 09 Apr 2014, Published online: 24 Jul 2014

Abstract

The uneventful progression through the cell cycle is closely associated with the rhythm set by the circadian clock machinery, with the S-phase of the cell cycle typically occurring at night. Presence of unrepaired DNA damage may reset the phase of the circadian clock, providing opportunities for damage assessment, repair and/or the induction of pro-apoptotic pathways. The core proteins of the circadian clock regulate directly or indirectly a significant number of genes coding for proteins involved in checkpoint transition, cell proliferation and programmed cell death. Disruption of the circadian rhythm may increase the risk for some multifactorial diseases and conditions, including glucose intolerance, cardiovascular disease and various common cancers. In patients with cancer, chronic circadian misalignment may stimulate the growth of tumours and may modify the outcomes of anticancer therapy. Knowledge about the role of physiological rhythms in human disease may contribute to the field of individualized medicine, specifically, in risk assessment and prognostication of the outcomes in patients with multifactorial disease.

Abbreviations

NER:=

Nucleotide excision repair

Per:=

Period

CNS:=

Central nervous system

Cry:=

Cryptochrome

Introduction

Several drosophila, mouse and rat models with altered or disrupted periodicity regulator genes have already been created.[Citation1–5] The resulting phenotypes may significantly vary with regard to capacity to maintain rhythmicity in absence of entraining cues (from ‘rhythmic albeit phase-shifted’ to ‘completely arrhythmic’) and length of cycle (22–28 hours).[Citation6] Some of the mouse models with disrupted core periodicity genes may exhibit accelerated aging phenotypes of varying severity and cancer proneness.

Clock mouse mutants

Mutations in the Clock gene in mice alters the duration of the diurnal cycle in animals housed in constant darkness, although the rhythm is not completely lost.[Citation7] Homozygous Clock mutants may exhibit deregulation of feeding rhythmicity and reduced energy expenditure. They are prone to overeating and rapidly develop obesity, hyperglycemia, hypoinsulinemia and hyperlipidemia.[Citation8] Clock-deficient mice have shorter-than-normal lifespan and somewhat increased incidence of age-related cataract and dermatitis.[Citation9] The reproductive fitness was moderately decreased both in Clock-deficient mice and in carriers of the Δ19 mutation in the Clock gene (causing ‘skipping’ of exon 19).[Citation10,11] Clock-deficient mice may exhibit increased activity levels, increased sensitization to alcohol and cocaine and increased drug reward compared to wild-type mice,[Citation12] leading to the supposition that the Clock protein was involved in the regulation of the dopaminergic system in mammals. The incidence of cancer in Clock-deficient mice has not been found to be increased when compared to non-mutant animals.[Citation13]

Bmal1 mouse mutants

The loss of the Bmal1 gene in mice results in immediate loss of circadian rhythmicity when the animals are housed in constant darkness.[Citation6] Bmal1 mutant mice also exhibit impaired glucose tolerance and decreased insulin secretion that tended to become worse with advancing age.[Citation14] Bmal1-deficient mice are sterile and have severely shortened lifespan (27 weeks on the average, compared to 70–150 weeks in normal mice), have lower body weight compared to wild-type mice and also exhibit various traits associated with premature aging such as early development of cataracts, arthropathy, ectopic calcification and loss of muscle and subcutaneous fat.[Citation15–17] Data about cancer-proneness in homozygous Bmal1 mutant mice is unreliable, as the phenotype of accelerated aging causes early death. Some of the homozygote mice exhibited hyperplasia of the salivary glands and several per cent of them developed lymphoma after gamma-irradiation, although their lifespan was short either way. Heterozygous Bmal1-knockout mice exhibit increased cancer-proneness, spontaneous as well as after genotoxic challenge (ionizing radiation).[Citation17]

Per and Cry mouse mutants

In mouse models, genes coding for both Cry proteins must be disrupted to produce a phenotype of deregulation of circadian rhythms, as normal functioning of the product of the one gene may partially substitute for the other.[Citation3,Citation18] Only mice with combined Cry1/Cry2 homozygous knockouts instantly lose the diurnal rhythm when housed in complete darkness, whereas mice lacking the expression of only one of the two proteins exhibit longer or shorter cycle when housed in complete darkness.[Citation3] Mice with targeted disruption of the Per3 gene exhibit only subtle disturbances of the circadian clock.[Citation19]

The circadian clock is adjustable by food cues. Mice deficient in Per2 are unable to predict and anticipate the approximate time when food is normally available.[Citation19,20] Mice with mutant Npas2 or deficient for Cry1 and Cry2 are impaired in food-associated entrainment of the circadian clock.[Citation20]

Cry1 homozygous knockout mice, double Cry1/Cry2 knockout mice and Per2 mutant homozygous mutant mice exhibit increased rates of spontaneous tumours and tumours developing after genotoxic challenge (ionizing radiation).[Citation17,Citation21,Citation22,23] The circadian pattern of expression of some genes coding for proteins involved in the control of the progression in the cell cycle (c-Myc, Cyclin D1, Cyclin A, Mdm-2) is grossly deregulated in mice with mutant Per2.[Citation22,23]

Cry1/Cry2 deficient double mutant mice that also carry inactivated Tp53 gene copies exhibit longer cancer-free survival and somewhat extended lifespan than p53-deficient mice without Cry mutations, although both groups virtually never reach the lifespan of normal mice.[Citation24] This is believed to be associated with increased propensity toward apoptosis by the p53-independent mechanism, conferred by deficiency of Cry proteins.[Citation24]

Human phenotypes associated with carriership of mutations or polymorphisms in clock machinery genes

Accelerated aging is a prominent feature in the animal models of deficiency or modification of core clock genes. It is therefore possible that the oscillating circadian clock may be an adjusting factor for the general molecular clock of aging. Aging is currently viewed as a preprogrammed mechanism, rather than a simple product of wear and tear of tissues and organs.[Citation25] The rate of attrition of telomere ends is currently viewed as a timing mechanism of the unidirectional (hourglass) type.[Citation26] For single cells, telomere attrition rate is a marker for the cell's proximity to replicative senescence. On a higher level, however, telomere length and rate of attrition of telomere ends provide a basis for the assessment of the rate of aging of tissues, organs and organisms. Shortening of telomere ends as a designated mechanism for induction of aging has been intensively studied, as it provides quantitative data that may be measured and compared between experiments.[Citation27,28] Research has already shown that the circadian pattern of expression of clock genes becomes markedly impaired in senescent cells.[Citation29,30] This, however, was shown to be remedied (albeit temporarily) by telomere lengthening. It has been proposed that the functioning of the oscillator clocks is dependent on the ‘time’ shown by the unidirectional clock of aging, with the peripheral clocks in aged tissues becoming less responsive to the signals sent by the master clock.[Citation29]

In humans, the presence of variant alleles of genes coding for proteins functioning in the circadian clock may cause sleep phase shift syndromes. These are usually quite benign and only very rarely interfere significantly with the normal life of the affected individual. The Ser662Gly mutation (rs121908635) in the PER2 gene is known to cause advanced sleep phase syndrome 1, manifested by the need to go to bed very early in the evening (6–8 pm) and spontaneous awakening very early in the morning (3–4 am).[Citation31] The ‘natural short sleeper’ human phenotype is associated with heterozygous carriership of the mutation Pro385Arg in the DEC2 gene.[Citation32,33] Carrier individuals require fewer hours of sleep per 24 h (on the average, 6 hours) to be completely rested than what is considered normal in the general population (7–9 h per 24 h). Presence of the DEC2 Pro385Arg mutation does not seem to be associated with any adverse effects.

One noncoding polymorphism in the 3′-UTR of the human CLOCK gene (a T-to-C transition, rs1801260) is associated with adult attention-deficit and hyperactivity disorder (ADHD).[Citation34]

Several inherited polymorphisms in core clock genes may be associated with increased risk for development of various tumours. The 311T>C single-nucleotide polymorphism in the human CLOCK1 gene have been associated with significantly increased susceptibility to colorectal carcinoma.[Citation35] The 5-repeat variant allele of the 4/5 repeat polymorphism in the human PER3 gene is associated with almost twofold increased risk for breast cancer in premenopausal women.[Citation36] The Ala394Thr polymorphism (rs2305160) in the coding sequence (specifically, in the PAS domain) of the human NPAS2 gene has also been found to increase the risk for non-Hodgkin's lymphoma.[Citation37]

Current research data show that the disruption of the circadian cycle may increase the risk for common diseases and conditions (diabetes, cancer, cardiovascular disease) and/or accelerate their progression. Of course, as in all diseases with multifactorial genesis, the presence of an additional risk factor does not mean that the individual will definitely develop the disease or condition, only that in its presence the risk is increased compared to the general population.[Citation38] Considering that the factors disrupting the circadian rhythm are very common, especially in industrialized countries (light at night, insufficient lighting during the day, evening and night shift work and/or rotating shifts, major meal for the day consumed in the evening hours, frequent transmeridional flights, etc.), it could be expected that the health impact would continue to increase in the future. Lifestyle and jobs including disruption of the day-night rhythm (specifically, night shift work) may be associated with increased incidence of colorectal carcinoma, breast, lung and prostate cancer in man.[Citation39–43] The majority of the studies on the impact of disruption of circadian rhythms on human health were conducted in female controls (predominantly nurses), therefore, one cannot exclude gender-specific differences (at least for some cancers). There is also the fact that nurses, as health workers, were motivated for continuing participation in these studies. The attitude of the participating individuals towards any medical study (and, especially, studies related to cancer medicine) may seriously affect the study, in terms of attrition rate, reliability of the personal data provided by the study objects, and the potential health benefits (opportunities to care for one's health in a more efficient manner – e.g. having more regular checkups).[Citation44] Medical personnel are, on the whole, more likely to stick to the study from the beginning to the end, fill the questionnaires accurately (because of pre-existing knowledge about the problem under study) and have their medical checkups regularly, allowing for early diagnosis and more precise risk assessment. In a 2007 press release the International Agency for research on Cancer (IARC) pronounced that ‘…Shiftwork that involves circadian disruption is probably carcinogenic to humans…’ [http://www.iarc.fr/en/media-centre/pr/2007/pr180.html]. The risk has been found to be especially high for those on rotating night shifts, not allowing for adaption of the circadian clock to the timing of the subjective day and the subjective night. The increased risk for lung cancer in night shift workers was, however, modifiable by environmental factors (specifically, smoking)[Citation42], and was not reported in all studied populations.[Citation45]

The association between disruption of the diurnal rhythm and breast cancer has been particularly well studied. It has been speculated that disrupted circadian rhythms (lack of natural (sun-spectrum) lighting or simply insufficient lighting during the day and/or artificial lighting at night) may be at least partially responsible for the increasing rates of breast cancer in industrialized countries. There have been reports about lower overall incidence of cancer in people with total visual blindness (could not perceive light at all) compared to the general population.[Citation46] The association was specifically strong for mammary gland cancer (over twofold risk reduction in individuals with total visual blindness than in the general population),[Citation47,48] but the risk for prostate cancer was also found to be lower among totally blind men.[Citation49] Since hormone release usually follows a circadian rhythm, and the majority of breast and prostate cancers are hormone-dependent, these results are not unexpected.

Disruption of circadian rhythms may modify the course of neoplastic disease. In recent studies, light at night promoted tumour growth in mouse models with breast cancer.[Citation50,51] In human patients with breast carcinoma, daily bedtime misalignment (i.e. misalignment between preferred bedtime and actual bedtime) was shown to be associated with more rapid cancer progression.[Citation52] It has even proposed that the risk for melanoma may be increased in individuals habitually exposed to excess light in the evening and night.[Citation53]

Mutations in circadian clock genes have been observed in tumour tissue – specifically, in colorectal cancer, breast cancer, prostate cancer and thyroid carcinoma.[Citation54–56] The expression of DEC1 is down-regulated or absent in >50% of oesophageal cancers in man.[Citation57] Disruption of the normal circadian cycle may be implemented on epigenetic level in tumour cells. One study of tumour tissues from breast cancer patients found hypermethylation on the promoters of PER1, PER2, CRY1 or BMAL1 genes in about 70% of the cases, compared to <50% methylation in non-cancerous tissues.[Citation54] In the same study, homogeneous (non-rhythmic) expression of PER2 or BMAL1 was significantly associated with lymph node metastasis and poorer prognosis for the patient. PER1 and PER2 genes are currently considered to be true tumour suppressor genes, as decreased expression of either (or both) has been reported in several types of human cancers (pancreatic cancer, renal cancer, head and neck cancers)[Citation58–60] and reduced PER1 and PER2 expression was recently shown to be an independent predictor of poorer prognosis in patients with gastric cancer.[Citation61] In 2009, it was proposed that the growth rate of breast cancer corresponded to the rhythm set by the circadian clock.[Citation62] Down-regulation of Per2 resulted in increases in the levels of Cyclin D and Cyclin E and accelerated tumour growth in vivo [Citation63] whereas induced overexpression of either Per1 or Per2 has been shown to inhibit the growth of cancer cells and increase their apoptotic rate.[Citation64] In patients with chronic lymphocytic leukaemia (CLL), the expression of BMAL1, PER1 and PER2 was found to be significantly down-regulated in comparison to healthy controls whereas the expression of the proto-oncogene c-MYC and Cyclin D1 was significantly up-regulated.[Citation65]

The expression profile of the core clock proteins may affect the outcomes in patients with cancer. Among patients with CLL, different CRY1 expression levels and CRY1/PER2 expression ratio were associated with different clinical course (and, respectively, very different outcomes), with epigenetically silenced CRY1 gene usually associated with indolent disease, needing treatment only in its very late phases, if ever.[Citation66,67] Thus, the levels of expression of circadian clock genes may be used as auxiliary markers in the prognostic panel in CLL.[Citation67–69] The overall survival of patients with colorectal cancer with high BMAL1 levels in the primary tumour was significantly longer (1.5 times) than that of patients with tumours with low BMAL1 levels; and the progression-free survival was more than two times higher in patients with high BMAL1 expression than that in patients with low expression.[Citation70] High levels of expression of the negative regulator of the core feedback loop CRY1 were associated with poorer overall survival in patients with colorectal cancer.[Citation71]

The levels of expression of the genes of the circadian core clock and the accessory proteins may predict sensitivity to anticancer therapies and/or provide information about the opportunities for therapeutic intervention. BMAL1 overexpression was shown to inhibit the growth of human colorectal cancer cell lines, increasing their sensitivity to genotoxic agents (oxaliplatin).[Citation70] It has been shown that Tim-depleted cancer cells may become sensitive to doxorubicin (a topoisomerase II inhibitor), making Tim1 a potential anticancer target in therapies based on ATM/ATR damage response pathway inhibition.[Citation72–74]

Currently, there is a massive research effort concentrated at studying the effects of timing of anticancer therapies around the circadian rhythm on the chances of achieving the maximal possible therapeutic effect (cancer chronotherapy).[Citation75] Most currently used anticancer agents work by infliction of DNA damage (genotoxicity). Their effect is strongest in rapidly dividing cells, as tumour cells are, but may affect the functioning of non-tumour cells with naturally rapid turnover as well. In principle, anticancer therapies are considered appropriate if they fulfil two requirements: (1) they produce a good objective response (tumour regression, slowing down the growth of the tumour) and (2) their use is coupled with minimal adverse effects for the patient. It is now agreed that anticancer therapy needs a considerable amount of customization to achieve the optimal balance of these two factors for the particular patient.[Citation76] Basically, in cancer chronotherapy, the application of the genotoxic agent/s is timed to the 24-h circadian variations (and, sometimes, to intradian (8–12 h) variations) in the mitotic index of cancer cells. Timing of genotoxic therapy to the specific time of the day when it is supposed to produce the greatest amount of damage to specific type of tumour cells may be expected to be associated with greater treatment response. At the same time, as the mitotic index of cancer and normal cells of the same tissue may peak at different times of the day, it could be expected that the associated toxicities for the normal tissues would be lower (least toxic times of chemotherapy). For example, a study among patients with ovarian cancer showed that the circadian peak in DNA synthesis for tumour cells was found between noon and 4 pm, which was ≈12 hours off the peak of DNA synthesis in non-tumour cells (usually occurring at night).[Citation77] Similarly, in patients with non-Hodgkin lymphoma, the peak of DNA synthesis in affected lymph nodes occurred at night, whereas the highest levels of DNA synthesis in healthy cells in the bone marrow were observed in the early hours of the afternoon.[Citation78] So far, the trials of cancer chronotherapy have shown that timing of genotoxic therapies within the circadian cycle produces significantly lower rates of severe adverse effects than ‘conventionally timed’ therapy.[Citation79–81] Indeed, undergoing cancer chronotherapy may mean significantly more treatment-related time spent in hospital, as the optimal time for drug administration may be late at night or very early in the morning, but since the outcomes may be objectively better than in conventionally timed therapy, there is every chance that chronotherapy for cancer may find broader applications in the near future. Obtaining data about lifestyle traits and habits possibly disrupting the circadian rhythm as well as testing for the presence of variants of the core clock genes may make a valuable addition to the panels of assessment of individual repair capacity, alongside polymorphisms in genes coding for proteins of DNA repair and maintenance of genome integrity and unscheduled DNA synthesis, especially in patients with cancer.[Citation28,Citation76,Citation82–84]

The circadian clock may play a role in the pathogenesis of multifactorial diseases and conditions other than cancer. For example, deregulated expression of Bmal1 and Cry1 was detected in animal models of traumatic brain injury.[Citation85] The sleep-wake cycle is grossly disturbed in patients with dementia of Alzheimer's type.[Citation86,87] It has been hypothesized that the regulation of the peripheral circadian clocks (e.g. in the cardiovascular system) may become more and more difficult with age.[Citation29] This impaired peripheral circadian rhythm may produce deregulated expression of tissue-specific clock-controlled genes, resulting in cardiovascular disease (e.g. plasminogen activator inhibitor-1, vascular endothelial growth factor and others).[Citation29,Citation88,Citation89]

It is well known that the occurrence of stroke usually peaks in the morning (roughly between 6 and 12 am).[Citation90] The same is valid for myocardial infarction and sudden cardiac death, where the risk for incidents is highest in the several hours after morning awakening.[Citation91] Neuronal susceptibility to ischemic events seems to follow a circadian pattern.[Citation92] In Per1-deficient-mice with cerebral ischaemia, higher rates of neuronal injury have been observed than in non-mutant ischaemic controls.[Citation93] CNS cells exhibit variations in the activation of apoptosis markers (expression of caspases-3, -8 and -9) in response to cerebral ischemia induced at different times of day.[Citation93,94] Ischemia is typically associated with increased levels of oxidative stress. It is possible that the disruption of the circadian rhythm associated with oxidative DNA damage may impair the functioning of the system for the assessment of the scale and scope of the damage, with the result that damaged cells in clock-disrupted models are more likely to be routed to the apoptotic pathway than in controls (where the same amount of DNA damage might have been assessed as ‘repairable’). Carriership of variant alleles associated with increased levels of unrepaired DNA damage and the associated propensity toward apoptosis in the vascular wall may contribute to the ‘morning peak’, and to the disruption of the circadian cycle commonly seen in stroke patients in the period immediately following the incident.[Citation95,96]

Circadian misalignment has been shown to increase insulin resistance and augment expression of inflammatory markers in diabetes type 2.[Citation97] The decline in beta-cell function seen in the progression of diabetes type 2 may be accelerated by the disruption of circadian rhythms and accelerate development of diabetes type 2.[Citation98,99]

Conclusions

Circadian clock dysfunction plays a role in the pathogenesis of many common multifactorial diseases and conditions, including glucose intolerance, cardiovascular disease and cancer. The core clock proteins may directly or indirectly modulate the expression of proteins functioning in the progression of the cell cycle. Disruption of the rhythm set by the internal clock may increase the risk for development of disease, aggravate the course of pre-existing conditions and modulate the outcomes of anticancer therapy. Knowledge about the mechanisms governing the maintenance of circadian rhythms and recovery of this rhythm after disruption may provide opportunities for informed lifestyle modification, more efficient therapeutic intervention and may assist in more individualized selection of anticancer therapies and schedules as well the prognostication of outcomes in patients with multifactorial diseases.

References

  • Sehgal A, Price JL, Man B, Young MW. Loss of circadian behavioral rhythms and per RNA oscillations in the drosophila mutant timeless. Science. 1994;263(5153):1603–1606.
  • Vitaterna MH, King DP, Chang A-M, Kornhauser JM, Lowrey PL, McDonald JD, Dove WF, Pinto LH, Turek FW, Takahashi JS. Mutagenesis and mapping of a mouse gene, Clock, essential for circadian behaviour. Science. 1994;264(5159):719–725.
  • van der Horst GT, Muijtjens M, Kobayashi K, Takano R, Kanno S, Takao M, de Wit J, Verkerk A, Eker AP, van Leenen D, Buijs R, Bootsma D, Hoeijmakers JH, Yasui A. Mammalian Cry1 and Cry2 are essential for maintenance of circadian rhythms. Nature. 1999;398(6728):627–630.
  • Okamura H, Miyake S, Sumi Y, Yamaguchi S, Yasui A, Muijtjens M, Hoeijmakers JHJ, van der Horst GTJ. Photic induction of mPer1 and mPer2 in Cry-deficient mice lacking a biological clock. Science.1999;286(5449):2531–2534.
  • Bunger MK, Wilsbacher LD, Moran SM, Clendenin C, Radcliffe LA, Hogenesch JB, Simon MC, Takahashi JS, Bradfield CA. Mop3 is an essential component of the master circadian pacemaker in mammals. Cell. 2000;103:1009–1017.
  • Yu EA, Weaver DR. Disrupting the circadian clock: gene-specific effects on aging, cancer, and other phenotypes. Aging (Albany, NY). 2011;3(5):479–493.
  • King DP, Zhao Y, Sangoram AM, Wilsbacher LD, Tanaka M, Antoch MP, Steeves TDL, Vitaterna MH, Kornhauser JM, Lowrey PL, Turek FW, Takahashi JS. Positional cloning of the mouse circadian clock gene. Cell. 1997;89:641–653.
  • Turek FW, Joshu C, Kohsaka A, Lin E, Ivanova G, McDearmon E, Laposky A, Losee-Olson S, Easton A, Jensen DR, Eckel RH, Takahashi JS, Bass J. Obesity and metabolic syndrome in circadian clock mutant mice. Science. 2005;308(5724):1043–1045.
  • Dubrovsky YV, Samsa WE, Kondratov RV. Deficiency of circadian protein clock reduces lifespan and increases age-related cataract development in mice. Aging (Albany NY). 2010; 2(12):936–944.
  • Dolatshad H, Campbell EA, O’Hara L, Maywood ES, Hastings MH, Johnson MH. Developmental and reproductive performance in circadian mutant mice. Hum Reprod. 2006;21:68–79.
  • Kennaway DJ, Boden MJ, Voultsios A. Reproductive performance in female clock Delta19 mutant mice. Reprod Fertil Dev. 2004;16:801–810.
  • McClung CA, Sidiropoulou K, Vitaterna M, Takahashi JS, White FJ, Cooper DI, Nestler EJ. Regulation of dopaminergic transmission and cocaine reward by the Clock gene. Proc Natl Acad Sci USA. 2005;102:9377–9381.
  • Antoch MP, Gorbacheva VY, Vykhovanets O, Toshkov IA, Kondratov RV, Kondratova AA, Lee C, Nikitin AY. Disruption of the circadian clock due to the clock mutation has discrete effects on aging and carcinogenesis. Cell Cycle. 2008;7:1197–1204.
  • Marcheva B, Ramsey KM, Buhr ED, Kobayashi Y, Su H, Ko CH, Ivanova G, Omura C, Mo S, Vitaterna MH, Lopez JP, Philipson LH, Bradfield CA, Crosby SD, JeBailey L, Wang X, Takahashi JS, Bass J. Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature. 2010;466(7306):627–631.
  • Kondratov RV, Kondratova AA, Lee C, Gorbacheva VY, Chernov MV, Antoch MP. Post-translational regulation of circadian transcriptional CLOCK(NPAS2)/BMAL1 complex by CRYPTOCHROMES. Cell Cycle. 2006;5(8):890–895.
  • Alvarez JD, Hansen A, Ord T, Bebas P, Chappell PE, Giebultowicz JM. Williams C Moss S, Sehgal A. The circadian clock protein BMAL1 is necessary for fertility and proper testosterone production in mice. J Biol Rhythms. 2008;23(1):26–36.
  • Lee S, Donehower LA, Herron AJ, Moore DD, Fu L. Disrupting circadian homeostasis of sympathetic signaling promotes tumor development in mice. PLoS One. 2010;5:e10995.
  • Vitaterna MH, Selby CP, Todo T, Niwa H, Thompson C, Fruechte EM, Hitomi K, Thresher RJ, Ishikawa T, Miyazaki J, Takahashi JS, Sancar A. Differential regulation of mammalian period genes and circadian rhythmicity by cryptochromes 1 and 2. Proc Natl Acad Sci USA. 1999; 96(21):12114–12119.
  • Shearman LP, Jin X, Lee C, Reppert SM, Weaver DR. Targeted disruption of the mPer3 gene: subtle effects on circadian clock function. Mol Cell Biol. 2000;20:6269–6275.
  • Feillet CA, Ripperger JA, Magnone MC, Dulloo A, Albrecht U, Challet E. Lack of food anticipation in Per2 mutant mice. Curr Biol. 2006;16(20):2016–2022.
  • Feillet CA, Albrecht U, Challet EJ. ‘Feeding time’ for the brain: a matter of clocks. Physiol Paris. 2006;100(5–6):252–260.
  • Lee CC. The circadian clock and tumor suppression by mammalian period genes. Methods Enzymol. 2005;393,852–861.
  • Chen-Goodspeed M, Lee CC. Tumor suppression and circadian function. J Biol Rhythms. 2007;22(4):291–298.
  • Ozturk N, Lee JH, Gaddameedhi S, Sancar A. Loss of cryptochrome reduces cancer risk in p53 mutant mice. Proc Natl Acad Sci USA. 2009;106(8):2841–2846.
  • Khalil HS, Petkova R, Zhelev N. Differential genetic advantages in youth and in aging, or how to die healthy. Biotechnol Biotechnol Equipment. 2012;26(1):2703–2711.
  • Rensing L, Meyer-Grahle U, Ruoff P. Biological timing and the clock metaphor: oscillatory and hourglass mechanisms. Chronobiol Int. 2001;18(3):329–369.
  • Petkova R, Chicheva Z, Chakarov S. Measuring telomere length – from ends to means. Biotechnol Biotechnol Equipment. 2011;25(4):2576–2582.
  • Chicheva Z, Chelenkova P, Petkova R, Chakarov S. Children of the Sun, children of the Moon – a mini-panel for assessment of inter-individual variation between the capacity of healthy individuals to repair everyday genotoxic insults. Biotechnol Biotechnol Equipment. 2012;26(4):3142–3147.
  • Kunieda T, Minamino T, Katsuno T, Tateno K, Nishi J, Miyauchi H, Orimo M, Okada S, Komuro I. Cellular senescence impairs circadian expression of clock genes in vitro and in vivo. Circ Res. 2006;98(4):532–539.
  • Qu Y, Mao M, Li X, Liu Y, Ding J, Jiang Z, Wan C, Zhang L, Wang Z, Mu D. Telomerase reconstitution contributes to resetting of circadian rhythm in fibroblasts. Mol Cell Biochem. 2008;313(1–2):11–18.
  • Toh KL, Jones CR, He Y, Eide EJ, Hinz WA, Virshup DM, Ptacek LJ, Fu Y-H. An hPer2 phosphorylation site mutation in familial advanced sleep phase syndrome. Science. 2001;291(5506):1040–1043.
  • Honma S, Kawamoto T, Takagi Y, Fujimoto K, Sato F, Noshiro M, Kato Y, Honma K. Dec1 and Dec2 are regulators of the mammalian molecular clock. Nature. 2002;419(6909):841–844.
  • He Y, Jones CR, Fujiki N, Xu Y, Guo B, Holder JL Jr, Rossner MJ, Nishino S, Fu Y-H. The transcriptional repressor DEC2 regulates sleep length in mammals. Science. 2009;325(5942):866–870.
  • Kissling C, Retz W, Wiemann S, Coogan AN, Clement RM, Hünnerkopf R, Conner AC, Freitag CM, Rösler M, Thome J. A polymorphism at the 3′-untranslated region of the CLOCK gene is associated with adult attention-deficit hyperactivity disorder. Am J Med Genet B Neuropsychiatric Genet. 2008;147(3):333–338.
  • Karantanos T, Theodoropoulos G, Gazouli M, Vaiopoulou A, Karantanou C, Stravopodis DJ, Bramis K, Lymperi M, Pektasidis D. Association of the clock genes polymorphisms with colorectal cancer susceptibility. J Surg Oncol. 2013;108(8):563–567.
  • Zhu Y, Brown HN, Zhang Y, Stevens RG, Zheng T. Period3 structural variation: a circadian biomarker associated with breast cancer in young women. Cancer Epidemiol Biomarkers Prev. 2005;14(1):268–270.
  • Zhu Y, Leaderer D, Guss C, Brown HN, Zhang Y, Boyle P, Stevens RG, Hoffman A, Qin Q, Han X, Zheng T. Ala394Thr polymorphism in the clock gene NPAS2: a circadian modifier for the risk of non-Hodgkin's lymphoma. Int J Cancer. 2007;120(2):432–435.
  • Petkova R, Chakarov S, Ganev V. Genetic bases for predisposition to common multifactorial disease in man. Part I. Biotechnol Biotechnol Equipment. 2007; 21(3):286–293.
  • Schernhammer ES, Kroenke CH, Laden F, Hankinson SE. Night work and risk of breast cancer. Epidemiology. 2006;17(1):108–111.
  • Hansen J, Lassen CF. Nested case-control study of night shift work and breast cancer risk among women in the Danish military. Occup Environ Med. 2012;69(8):551–556.
  • Flynn-Evans EE, Mucci L, Stevens RG, Lockley SW. Shiftwork and prostate-specific antigen in the National Health and Nutrition Examination Survey. J Natl Cancer Inst. 2013;105(17):1292–1297.
  • Schernhammer ES, Feskanich D, Liang G, Han J. Rotating night-shift work and lung cancer risk among female nurses in the United States. Am J Epidemiol. 2013;178(9):1434–1441.
  • Sigurdardottir LG, Valdimarsdottir UA, Mucci LA, Fall K, Rider JR, Schernhammer E, Czeisler CA, Launer L, Harris T, Stampfer MJ, Gudnason V, Lockley SW. Sleep disruption among older men and risk of prostate cancer. Cancer Epidemiol Biomarkers Prev. 2013;22(5):872–879.
  • Petkova R, Chakarov S, Ganev V. Genetic bases for predisposition to common multifactorial disease in man. Part II. Biotechnol Biotechnol Equipment. 2007;21(4):385–392.
  • Kloog I, Haim A, Stevens RG, Barchana M, Portnov BA. Light at night co-distributes with incident breast but not lung cancer in the female population of Israel. Chronobiol Int. 2008;25(1):65–81.
  • Feychting M, Osterlund B, Ahlbom A. Reduced cancer incidence among the blind. Epidemiology. 1998;9(5):490–494.
  • Kliukiene J, Tynes T, Andersen A. Risk of breast cancer among Norwegian women with visual impairment. Br J Cancer. 2001;84(3):397–399.
  • Flynn-Evans EE, Stevens RG, Tabandeh H, Schernhammer ES, Lockley S. Total visual blindness is protective against breast cancer. Cancer Causes Control. 2009;20(9):1753–1756.
  • Pukkala E, Ojamo M, Rudanko SL, Stevens RG, Verkasalo PK. Does incidence of breast cancer and prostate cancer decrease with increasing degree of visual impairment? Cancer Causes Control. 2006;17(4):573–576.
  • Savvidis C, Koutsilieris M. Circadian rhythm disruption in cancer biology. Mol Med. 2012;18:1249–1260.
  • Schwimmer H, Metzer A, Pilosof Y, Szyf M, Machnes ZM, Fares F, Harel O, Haim A. Light at night and melatonin have opposite effects on breast cancer tumors in mice assessed by growth rates and global DNA methylation. Chronobiol Int. 2014;31(1):144–150.
  • Hahm BJ, Jo B, Dhabhar FS, Palesh O, Aldridge-Gerry A, Bajestan SN, Neri E, Nouriani B, Spiegel D, Zeitzer JM. Bedtime misalignment and progression of breast cancer. Chronobiol Int. 2014;31(2):214–221.
  • Kvaskoff M, Weinstein P. Are some melanomas caused by artificial light? Med Hypotheses. 2010;75(3):305–311.
  • Kuo SJ, Chen ST, Yeh KT, Hou MF, Chang YS, Hsu NC, Chang JG. Disturbance of circadian gene expression in breast cancer. Virchows Arch. 2009;454(4):467–474.
  • Oshima T, Takenoshita S, Akaike M, Kunisaki C, Fujii S, Nozaki A, Numata K, Shiozawa M, Rino Y, Tanaka K, Masuda M, Imada T. Expression of circadian genes correlates with liver metastasis and outcomes in colorectal cancer. Oncol Rep. 2011;25(5):1439–1446.
  • Mannic T, Meyer P, Triponez F, Pusztaszeri M, Le Martelot G, Mariani O, Schmitter D, Sage D, Philippe J, Dibner C. Circadian clock characteristics are altered in human thyroid malignant nodules. J Clin Endocrinol Metab. 2013;98(11):4446–4456.
  • Nishiwaki T, Daigo Y, Kawasoe T, Nakamura Y. Isolation and mutational analysis of a novel human cDNA, DEC1 (deleted in esophageal cancer 1), derived from the tumor suppressor locus in 9q32. Genes Chromosomes Cancer. 2000;27:169–176.
  • Hsu CM, Lin SF, Lu CT, Lin PM, Yang MY. Altered expression of circadian clock genes in head and neck squamous cell carcinoma. Tumour Biol. 2012;33(1):149–155.
  • Mazzoccoli G, Piepoli A, Carella M, Panza A, Pazienza V, Benegiamo G, Palumbo O, Ranieri E. Altered expression of the clock gene machinery in kidney cancer patients. Biomed Pharmacother. 2012;66(3):175–179.
  • Relles D, Sendecki J, Chipitsyna G, Hyslop T, Yeo CJ, Arafat HA. Circadian gene expression and clinicopathologic correlates in pancreatic cancer. J Gastrointest Surg. 2013;17(3):443–450.
  • Zhao H, Zeng ZL, Yang J, Jin Y, Qiu MZ, Hu XY, Han J, Liu KY, Liao JW, Xu RH, Zou QF. Prognostic relevance of Period1 (Per1) and Period2 (Per2) expression in human gastric cancer. Int J Clin Exp Pathol. 2014;7(2):619–630.
  • Yang X, Wood PA, Oh EY, Du-Quiton J, Ansell CM, Hrushesky WJ. Down regulation of circadian clock gene Period 2 accelerates breast cancer growth by altering its daily growth rhythm. Breast Cancer Res Treat. 2009;117(2):423–431.
  • Yang X, Wood PA, Ansell CM, Quiton DF, Oh EY, Du-Quiton J, Hrushesky WJ. The circadian clock gene Per1 suppresses cancer cell proliferation and tumor growth at specific times of day. Chronobiol Int. 2009;26(7):1323–1339.
  • Miyazaki K, Wakabayashi M, Hara Y, Ishida N. Tumor growth suppression in vivo by overexpression of the circadian component, PER2. Genes Cells. 2010;15(4):351–358.
  • Rana S, Munawar M, Shahid A, Malik M, Ullah H, Fatima W, Mohsin S, Mahmood S. Deregulated expression of circadian clock and clock-controlled cell cycle genes in chronic lymphocytic leukemia. Mol Biol Rep. 2014;41(1):95–103.
  • Eisele L, Prinz R, Klein-Hitpass L, Nückel H, Lowinski K, Thomale J, Moeller LC, Dührsen U, Dürig J. Combined PER2 and CRY1 expression predicts outcome in chronic lymphocytic leukemia. Eur J Haematol. 2009;83(4):320–327.
  • Hanoun M, Eisele L, Suzuki M, Greally JM, Hüttmann A, Aydin S, Scholtysik R, Klein-Hitpass L, Dührsen U, Dürig J. Epigenetic silencing of the circadian clock gene CRY1 is associated with an indolent clinical course in chronic lymphocytic leukemia. PLoS One. 2012;7(3):e34347.
  • Winkler D, Döhner H, Stilgenbauer S. Genetics, gene expression, and targeted therapies in chronic lymphocytic leukemia. Curr Drug Targets. 2006;7(10):1313–1327.
  • Tummala H, Goltsov A, Khalil HS, Sproul A, Scott F, Mitev V, Zhelev N. Advocating the need of a systems biology approach for personalised prognosis and treatment of B-CLL patients. Biodiscovery. 2012;6:4.
  • Zeng ZL, Luo HY, Yang J, Wu WJ, Chen DL, Huang P, Xu RH. Overexpression of the circadian clock gene bmal1 increases sensitivity to oxaliplatin in colorectal cancer. Clin Cancer Res. 2014;20(4):1042–1052.
  • Yu H, Meng X, Wu J, Pan C, Ying X, Zhou Y, Liu R, Huang W. Cryptochrome 1 overexpression correlates with tumor progression and poor prognosis in patients with colorectal cancer. PLoS One. 2013;8(4):e61679.
  • Khalil HS, Tummala H, Chakarov S, Zhelev N, Lane DP. Targeting ATM pathway for therapeutic intervention in cancer. Biodiscovery. 2012;1:3.
  • Yang X, Wood PA, Hrushesky WJ. Mammalian TIMELESS is required for ATM-dependent CHK2 activation and G2/M checkpoint control. J Biol Chem. 2010;285(5):3030–3034.
  • Idowu MA, Khalil HS, Bown JL, Zhelev N. Reverse engineering of drug induced DNA damage response signalling pathway reveals dual outcomes of ATM kinase inhibition. Biodiscovery. 2013;9:4.
  • Mormont MC, Levi F. Cancer chronotherapy: principles, applications, and perspectives. Cancer. 2003;97(1):155–169.
  • Petkova R, Chelenkova P, Georgieva E, Chakarov S. What's your poison? Impact of individual repair capacity on the outcomes of genotoxic therapies in cancer. Part I – role of individual repair capacity in the constitution of risk for late-onset multifactorial disease. Biotechnol Biotechnol Equipment. 2013;27(6):4208–4216.
  • Klevecz R, Braly P. Circadian and ultradian cytokinetic rhythms of spontaneous human cancer. Ann NY Acad Sci. 1991;618:257–276.
  • Smaaland R, Lote K, Sothern RB, Laerum OD. DNA synthesis and ploidy in non-Hodgkin's lymphomas demonstrate intrapatient variation depending on circadian stage of cell sampling. Cancer Res. 1993;53(13):3129–3138.
  • Tampellini M, Filipski E, Liu XH, Lemaigre G, Li XM, Vrignaud P, François E, Bissery MC, Lévi F. Docetaxel chronopharmacology in mice. Cancer Res. 1998;58(17):3896–3904.
  • Granda TG, D’Attino RM, Filipski E, Vrignaud P, Garufi C, Terzoli E, Bissery MC, Lévi F. Circadian optimisation of irinotecan and oxaliplatin efficacy in mice with Glasgow osteosarcoma. Br J Cancer. 2002;86(6):999–1005.
  • Granda TG, Lévi F. Tumor-based rhythms of anticancer efficacy in experimental models. Chronobiol Int. 2002;19:21–41.
  • Chakarov S, Stoilov P, Alexandrov A, Russev G. Repair pattern in the beta-globin gene cluster of human fibroblasts after ultraviolet irradiation. Eur J Biochem. 1997;248:669–675.
  • Hastings MH, Reddy AB, Maywood ES. A clockwork web: circadian timing in brain and periphery, in health and disease. Nat Rev Neurosci. 2003;4(8):649–666.
  • Chelenkova P, Petkova R, D’ Ascanio I, Zhelev N, Chakarov S. In sickness and in health: a set of markers for individual repair capacity in risk assessment, monitoring and prognosis of human disease. Curr Opin Biotechnol. 2013;24(suppl. 1):S105.
  • Boone DR, Sell SL, Micci MA, Crookshanks JM, Parsley M, Uchida T, Prough DS, DeWitt DS, Hellmich HL. Traumatic brain injury-induced dysregulation of the circadian clock. PLoS One. 2012;7(10):e46204.
  • Hatfield CF, Herbert J, van Someren EJ, Hodges JR, Hastings MH. Disrupted daily activity/rest cycles in relation to daily cortisol rhythms of home-dwelling patients with early Alzheimer's dementia. Brain. 2004;127(Pt 5):1061–1074.
  • Wu YH, Fischer DF, Kalsbeek A, Garidou-Boof ML, van der Vliet J, van Heijningen C, Liu RY, Zhou JN, Swaab DF. Pineal clock gene oscillation is disturbed in Alzheimer's disease, due to functional disconnection from the master clock. FASEB J. 2006;20(11):1874–1876.
  • Maemura K, de la Monte SM, Chin MT, Layne MD, Hsieh CM, Yet SF, Perrella MA, Lee ME. CLIF, a novel cycle-like factor, regulates the circadian oscillation of plasminogen activator inhibitor-1 gene expression. J Biol Chem. 2000;275:36847–36851.
  • Koyanagi S, Kuramoto Y, Nakagawa H, Aramaki H, Ohdo S, Soeda S, Shimeno H. A molecular mechanism regulating circadian expression of vascular endothelial growth factor in tumor cells. Cancer Res. 2003;63:7277–7283.
  • Elliott WJ. Circadian variation in the timing of stroke onset: a meta-analysis. Stroke. 1998;29(5):992–996.
  • Willich SN. Circadian variation and triggering of cardiovascular events. Vasc Med. 1999;4(1):41–49.
  • Karmarkar SW, Tischkau SA. Influences of the circadian clock on neuronal susceptibility to excitotoxicity. Front Physiol. 2013;4:313.
  • Wiebking N, Maronde E, Rami A. Increased neuronal injury in clock gene Per-1 deficient-mice after cerebral ischemia. Curr Neurovasc Res. 2013;10(2):112–125.
  • Tischkau SA, Cohen JA, Stark JT, Gross DR, Bottum KM. Time-of-day affects expression of hippocampal markers for ischemic damage induced by global ischemia. Exp Neurol. 2007;208(2):314–322.
  • Cavalcanti PR, Campos TF, Araüjo JF. Circadian and homeostatic changes of sleep-wake and quality of life in stroke: implications for neurorehabilitation. Neurorehabilitation. 2013;32(2):337–343.
  • Chelenkova P, Petkova R, Chamova T, Zheliazkova-Glaveeva S, Tournev I, Chakarov S. Homozygous carriership of the wildtype allele of the XPCins83 polymorphism is an independent protective factor against cerebrovascular incidents in the Bulgarian population. Compt Rend Acad Bulgarian Sci. 2014;67(2):263–268.
  • Leproult R, Holmbäck U, Van Cauter E. Circadian misalignment augments markers of insulin resistance and inflammation, independently of sleep loss. Diabetes. 2014;63(6).
  • Gale JE, Cox HI, Qian J, Block GD, Colwell CS, Matveyenko AV. Disruption of circadian rhythms accelerates development of diabetes through pancreatic beta-cell loss and dysfunction. J Biol Rhythms. 2011;26(5):423–433.
  • Rakshit K, Thomas AP, Matveyenko AV. Does disruption of circadian rhythms contribute to Beta-cell failure in type 2 diabetes? Curr Diab Rep. 2014;14(4):474.