Pharmacopsychiatry 2013; 46(S 01): S22-S29
DOI: 10.1055/s-0033-1337923
Original Paper
© Georg Thieme Verlag KG Stuttgart · New York

Circadian Rhythms Investigated on the Cellular and Molecular Levels

P. J. Gebicke-Haerter
1   Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
,
L. V. Pildaín
2   Department of Applied Mathematics and Center for Modeling and Simulation in the e Biosciences, University of ­Heidelberg, Heidelberg, Germany
,
F. Matthäus
2   Department of Applied Mathematics and Center for Modeling and Simulation in the e Biosciences, University of ­Heidelberg, Heidelberg, Germany
,
A. Schmitt
3   Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University of Munich, Munich, Germany
,
P. Falkai
3   Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University of Munich, Munich, Germany
› Institutsangaben
Weitere Informationen

Publikationsverlauf

Publikationsdatum:
18. April 2013 (online)

Abstract

Investigations on circadian rhythms have markedly advanced our understanding of health and disease with the advent of high-throughput technologies like microarrays and epigenetic profiling. They elucidated the multi-level behaviour of interactive oscillations from molecules to neuronal networks and eventually to processes of learning and memory in an impressive manner. The small-world topology of synchronized firing through neuron-neuron and neuron-glia gap junctions is discussed as a mathematical approach to these intensively studied issues. It has become evident that, apart from some disorders caused by gene mutations, the majority of disorders originating from disturbances of rhythms arise from environmental influences and epigenetic changes. In this context, it was mandatory to think of and devise experiments on temporary scales, which exponentially increased the volumes of data obtained from time-series and rapidly became prohibitive of manual inspection. Therefore, more and more sophisticated mathematical algorithms have been developed to identify rhythmic expression of genes and to find coexpression by their clustering. It is expected that disturbed rhythmic behaviour in mental disorders is reflected in altered oscillatory behaviour of gene expression.

 
  • References

  • 1 Sato TK, Yamada RG, Ukai H et al. Feedback repression is required for mammalian circadian clock function. Nat Genet 2006; 38: 312-319
  • 2 Zhang EE, Kay SA. Clocks not winding down: unravelling circadian networks. Nat Rev Mol Cell Biol 2010; 11: 764-776
  • 3 Wood PA, Yang X, Hrushesky WJ. Clock genes and cancer. Integr Cancer Ther 2009; 8: 303-308
  • 4 Sukumaran S, Almon RR, DuBois DC et al. Circadian rhythms in gene expression: Relationship to physiology, disease, drug disposition and drug action. Adv Drug Deliv Rev 2010; 62: 904-917
  • 5 Barnard AR, Nolan PM. When clocks go bad: neurobehavioural consequences of disrupted circadian timing. PLoS Genet 2008; 4: e1000040
  • 6 Liu HC, Hu CJ, Tang YC et al. A pilot study for circadian gene disturbance in dementia patients. Neurosci Lett 2008; 435: 229-233
  • 7 Ueda HR, Hayashi S, Chen W et al. System-level identification of transcriptional circuits underlying mammalian circadian clocks. Nat Genet 2005; 37: 187-192
  • 8 Baggs JE, Hogenesch JB. Genomics and systems approaches in the mammalian circadian clock. Curr Opin Genet Dev 2010; 20: 581-587
  • 9 Schibler U. The daily timing of gene expression and physiology in mammals. Dialogues Clin Neurosci 2007; 9: 257-272
  • 10 Hatanaka F, Matsubara C, Myung J et al. Genome-wide profiling of the core clock protein BMAL1 targets reveals a strict relationship with metabolism. Mol Cell Biol 2010; 30: 5636-5648
  • 11 Matsunaga N, Ikeda M, Takiguchi T et al. The molecular mechanism regulating 24-hour rhythm of CYP2E1 expression in the mouse liver. Hepatology 2008; 48: 240-251
  • 12 Crosio C, Cermakian N, Allis CD et al. Light induces chromatin modification in cells of the mammalian circadian clock. Nat Neurosci 2000; 3: 1241-1247
  • 13 Quintero JE, Kuhlman SJ, McMahon DG. The biological clock nucleus: a multiphasic oscillator network regulated by light. J Neurosci 2003; 23: 8070-8076
  • 14 Rensing L, Ruoff P. Temperature effect on entrainment, phase shifting, and amplitude of circadian clocks and its molecular bases. Chronobiol Int 2002; 19: 807-864
  • 15 Kino T, Chrousos GP. Acetylation-mediated epigenetic regulation of glucocorticoid receptor activity: circadian rhythm-associated alterations of glucocorticoid actions in target tissues. Mol Cell Endocrinol 2011; 336: 23-30
  • 16 Leonardson AS, Zhu J, Chen Y et al. The effect of food intake on gene expression in human peripheral blood. Hum Mol Genet 2010; 19: 159-169
  • 17 Gebicke-Haerter PJ. Epigenetics of schizophrenia. Pharmacopsychiatry 2012; 45 (Suppl. 1) S42-S48
  • 18 Bass J. Circadian topology of metabolism. Nature 2012; 491: 348-356
  • 19 Ralph MR, Foster RG, Davis FC et al. Transplanted suprachiasmatic nucleus determines circadian period. Science 1990; 247: 975-978
  • 20 Welsh DK, Logothetis DE, Meister M et al. Individual neurons dissociated from rat suprachiasmatic nucleus express independently phased circadian firing rhythms. Neuron 1995; 14: 697-706
  • 21 Pando MP, Morse D, Cermakian N et al. Phenotypic rescue of a peripheral clock genetic defect via SCN hierarchical dominance. Cell 2002; 110: 107-117
  • 22 Lin L, Faraco J, Li R et al. The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene. Cell 1999; 98: 365-376
  • 23 Hungs M, Fan J, Lin L et al. Identification and functional analysis of mutations in the hypocretin (orexin) genes of narcoleptic canines. Genome Res 2001; 11: 531-539
  • 24 Xu Y, Padiath QS, Shapiro RE et al. Functional consequences of a CKIdelta mutation causing familial advanced sleep phase syndrome. Nature 2005; 434: 640-644
  • 25 Toh KL, Jones CR, He Y et al. An hPer2 phosphorylation site mutation in familial advanced sleep phase syndrome. Science 2001; 291: 1040-1043
  • 26 Ebisawa T, Uchiyama M, Kajimura N et al. Association of structural polymorphisms in the human period3 gene with delayed sleep phase syndrome. EMBO Rep 2001; 2: 342-346
  • 27 Archer SN, Robilliard DL, Skene DJ et al. A length polymorphism in the circadian clock gene Per3 is linked to delayed sleep phase syndrome and extreme diurnal preference. Sleep 2003; 26: 413-415
  • 28 Kyriacou CP, Hastings MH. Circadian clocks: genes, sleep, and cognition. Trends Cogn Sci. 2010; 14: 259-267
  • 29 Ye R, Selby CP, Ozturk N et al. Biochemical analysis of the canonical model for the mammalian circadian clock. J Biol Chem 2011; 286: 25891-25902
  • 30 Wilkins AK, Barton PI, Tidor B. Per2 negative feedback loop sets the period in the mammalian circadian clock mechanism. PLoS Comput Biol 2007; 3: e242
  • 31 Hatanaka F, Matsubara C, Myung J et al. Genome-wide profiling of the core clock protein BMAL1 targets reveals a strict relationship with metabolism. Mol Cell Biol 2010; 30: 5636-5648
  • 32 Preitner N, Damiola F, Lopez-Molina L et al. The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell 2002; 110: 251-260
  • 33 Sato TK, Panda S, Miraglia LJ et al. A functional genomics strategy reveals Rora as a component of the mammalian circadian clock. Neuron 2004; 43: 527-537
  • 34 Etchegaray JP, Lee C, Wade PA et al. Rhythmic histone acetylation underlies transcription in the mammalian circadian clock. Nature 2003; 421: 177-182
  • 35 Doi M, Hirayama J, Sassone-Corsi P. Circadian regulator CLOCK is a histone acetyltransferase. Cell 2006; 125: 497-508
  • 36 Hirayama J, Sahar S, Grimaldi B et al. CLOCK-mediated acetylation of BMAL1 controls circadian function. Nature 2007; 450: 1086-1090
  • 37 Katada S, Sassone-Corsi P. The histone methyltransferase MLL1 permits the oscillation of circadian gene expression. Nat Struct Mol Biol 2010; 17: 1414-1421
  • 38 Nakahata Y, Kaluzova M, Grimaldi B et al. The NAD+-dependent deacetylase SIRT1 modulates CLOCK-mediated chromatin remodeling and circadian control. Cell 2008; 134: 329-340
  • 39 Asher G, Gatfield D, Stratmann M et al. SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell 2008; 134: 317-328
  • 40 Ripperger JA, Merrow M. Perfect timing: epigenetic regulation of the circadian clock. FEBS Lett 2011; 585: 1406-1411
  • 41 Masri S, Zocchi L, Katada S et al. The circadian clock transcriptional complex: metabolic feedback intersects with epigenetic control. Ann N Y Acad Sci 2012; 1264: 103-109
  • 42 Isobe Y, Hida H, Nishino H. Circadian rhythm of metabolic oscillation in suprachiasmatic nucleus depends on the mitochondrial oxidation state, reflected by cytochrome C oxidase and lactate dehydrogenase. J Neurosci Res 2011; 89: 929-935
  • 43 Asher G, Schibler U. A CLOCK-less clock. Trends Cell Biol 2006; 16: 547-549
  • 44 Asher G, Reinke H, Altmeyer M et al. Poly(ADP-ribose) polymerase 1 participates in the phase entrainment of circadian clocks to feeding. Cell 2010; 142: 943-953
  • 45 Kolthur-Seetharam U, Dantzer F, McBurney MW et al. Control of AIF-mediated cell death by the functional interplay of SIRT1 and PARP-1 in response to DNA damage. Cell Cycle 2006; 5: 873-877
  • 46 Feng D, Liu T, Sun Z et al. A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Science 2011; 331: 1315-1319
  • 47 Masri S, Sassone-Corsi P. The circadian clock: a framework linking metabolism, epigenetics and neuronal function. Nat Rev Neurosci 2013; 14: 69-75
  • 48 Lloyd AL, Lloyd D. Hypothesis: the central oscillator of the circadian clock is a controlled chaotic attractor. Biosystems 1993; 29: 77-85
  • 49 Masri S, Sassone-Corsi P. Plasticity and specificity of the circadian epigenome. Nat Neurosci 2010; 13: 1324-1329
  • 50 Yin L, Wu N, Curtin JC et al. Rev-erbalpha, a heme sensor that coordinates metabolic and circadian pathways. Science 2007; 318: 1786-1789
  • 51 Lukat-Rodgers GS, Correia C, Botuyan MV et al. Heme-based sensing by the mammalian circadian protein CLOCK. Inorg Chem 2010; 49: 6349-6365
  • 52 Hayasaka K, Kitanishi K, Igarashi J et al. Heme-binding characteristics of the isolated PAS-B domain of mouse Per2, a transcriptional regulatory factor associated with circadian rhythms. Biochim Biophys Acta 2011; 1814: 326-333
  • 53 McIntosh BE, Hogenesch JB, Bradfield CA. Mammalian Per-Arnt-Sim proteins in environmental adaptation. Annu Rev Physiol 2010; 72: 625-645
  • 54 Jiménez-Ortega V, Cardinali DP, Poliandri AH et al. 24-Hour rhythm in gene expression of nitric oxide synthase and heme-peroxidase in anterior pituitary of ethanol-fed rats. Neurosci Lett 2007; 425: 69-72
  • 55 Golombek DA, Agostino PV, Plano SA et al. Signaling in the mammalian circadian clock: the NO/cGMP pathway. Neurochem Int 2004; 45: 929-936
  • 56 Kaasik K, Lee CC. Reciprocal regulation of haem biosynthesis and the circadian clock in mammals. Nature 2004; 430: 467-471
  • 57 Meerlo P, Mistlberger RE, Jacobs BL et al. New neurons in the adult brain: the role of sleep and consequences of sleep loss. Sleep Med Rev 2009; 13: 187-194
  • 58 Wulff K, Gatti S, Wettstein JG et al. Opinion: Sleep and circadian rhythm disruption in psychiatric and neurodegenerative disease. Nat Rev Neurosci 2010; 11: 589-599
  • 59 Zsiros V, Maccaferri G. Electrical coupling between interneurons with different excitable properties in the stratum lacunosum-moleculare of the juvenile CA1 rat hippocampus. J Neurosci 2005; 25: 8686-8695
  • 60 Bauer EP, Paz R, Paré D. Gamma oscillations coordinate amygdalo-rhinal interactions during learning. J Neurosci 2007; 27: 9369-9379
  • 61 Pereira Jr A, Furlan FA. Astrocytes and human cognition: Modeling information integration and modulation of neuronal activity. Progr Neurobiol 2010; 92: 405-420
  • 62 Fukuda T, Kosaka T, Singer W et al. Gap junctions among dendrites of cortical GABAergic neurons establish a dense and widespread intercolumnar network. J Neurosci 2006; 26: 3434-3443
  • 63 Robertson JM. The astrocentric hypothesis: Proposed role of astrocytes in consciousness and memory formation. J Physiol (Paris) 2002; 96: 251-255
  • 64 Hirrlinger J, Hülsmann S, Kirchhoff F. Astroglial processes show spontaneous motility at active synaptic terminals in situ. Eur J Neurosci 2004; 20: 2235-2239
  • 65 Giuditta A, Chun JI, Eyman M et al. Local gene expressions in axons and nerve endings: The glia-neuron unit. Physiol Rev 2008; 88: 515-555
  • 66 Mitterauer BJ. Possible role of glia in cognitive impairment in schizophrenia. CNS Neurosci Ther 2011; 17: 333-344
  • 67 Mitterauer B. Loss of function of glial gap junctions may cause severe cognitive impairments in schizophrenia. Med Hypotheses 2009; 73: 393-397
  • 68 Behrendt RP. Dysregulation of thalamic sensory 'transmission' in schizophrenia: neurochemical vulnerability to hallucinations. J Psychopharmacol 2006; 20: 356-372
  • 69 Testu F, Clarisse R. Time-of-day and day-of-week effects on mnemonic performance. Chronobiol Int 1999; 16: 491-503
  • 70 Gerstner JR, Lyons LC, Wright Jr KP et al. Cycling behavior and memory formation. J Neurosci 2009; 29: 12824-12830
  • 71 Smit DJ, Boersma M, Schnack HG et al. The brain matures with stronger functional connectivity and decreased randomness of its network. PLoS One 2012; 7: e36896
  • 72 Watts DJ, Strogatz SH. Collective dynamics of ‘small-world’ networks. Nature 1998; 393: 440-442
  • 73 Bullmore E, Sporns O. Complex brain networks: graph theoretical analysis of structural and functional systems. Nat Rev Neurosci 2009; 10: 186-198
  • 74 Canolty RT, Ganguly K, Carmena JM. Task-dependent changes in cross-level coupling between single neurons and oscillatory activity in multiscale networks. PLoS Comput Biol 2012; 8: e1002809
  • 75 Siegel M, Warden MR, Miller EK. Phase-dependent neuronal coding of objects in short-term memory. Proc Natl Acad Sci USA 2009; 106: 21341-21346
  • 76 Koch H, Garcia 3rd AJ, Ramirez JM. Network reconfiguration and neuronal plasticity in rhythm-generating networks. Integr Comp Biol 2011; 51: 856-868
  • 77 Migita H, Morser J, Kawai K. Rev-erbalpha upregulates NF-kappaB-responsive genes in vascular smooth muscle cells. FEBS Lett 2004; 561: 69-74
  • 78 Yao Z, DuBois DC, Almon RR et al. Modeling circadian rhythms of glucocorticoid receptor and glutamine synthetase expression in rat skeletal muscle. Pharm Res 2006; 23: 670-679
  • 79 Rey G, Cesbron F, Rougemont J et al. Genome-wide and phase-specific DNA-binding rhythms of BMAL1 control circadian output functions in mouse liver. PLoS Biol 2011; 9: e1000595
  • 80 Yamamoto T, Nakahata Y, Soma H et al. Transcriptional oscillation of canonical clock genes in mouse peripheral tissues. BMC Molecular Biology 2004; 5: 18
  • 81 Kusanagi H, Hida A, Satoh K et al. Expression profiles of 10 circadian clock genes in human peripheral blood mononuclear cells. Neurosci Res 2008; 61: 136-142
  • 82 Takimoto M, Hamada A, Tomoda A et al. Daily expression of clock genes in whole blood cells in healthy subjects and a patient with circadian rhythm sleep disorder. Am J Physiol Regul Integr Comp Physiol 2005; 289: R1273-R1279
  • 83 Goodwin BC. Oscillatory behavior in enzymatic control processes. Adv Enzyme Regul 1965; 3: 425-438
  • 84 Goldbeter A. A model for circadian oscillations in the Drosophila period protein (PER). Proc Biol Sci 1995; 261: 319-324
  • 85 Leloup JC, Goldbeter A. A model for circadian rhythms in Drosophila incorporating the formation of a complex between the PER and TIM proteins. J Biol Rhythms 1998; 13: 70-87
  • 86 Goldbeter A. Computational approaches to cellular rhythms. Nature 2002; 420: 238-245
  • 87 Leloup JC, Goldbeter A. Toward a detailed computational model for the mammalian circadian clock. Proc Natl Acad Sci USA 2003; 100: 7051-7056
  • 88 Leloup JC, Goldbeter A. Modeling the mammalian circadian clock: sensitivity analysis and multiplicity of oscillatory mechanisms. J Theor Biol 2004; 230: 541-562
  • 89 Leloup JC, Goldbeter A. Modeling the circadian clock: from molecular mechanism to physiological disorders. Bioessays 2008; 30: 590-600
  • 90 Ukai-Tadenuma M, Kasukawa T, Ueda HR. Proof-by-synthesis of the transcriptional logic of mammalian circadian clocks. Nat Cell Biol 2008; 10: 1154-1163
  • 91 Gladkevich A, Kauffman HF, Korf J. Lymphocytes as a neural probe: potential for studying psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2004; 28: 559-576
  • 92 Zhu J, Chen Y, Leonardson AS et al. Characterizing dynamic changes in the human blood transcriptional network. PLoS Comput Biol 2010; 6: e1000671
  • 93 Hayashi M, Shimba S, Tezuka M. Characterization of the molecular clock in mouse peritoneal macrophages. Biol Pharm Bull 2007; 30: 621-626
  • 94 Arjona A, Sarkar DK. Circadian oscillations of clock genes, cytolytic factors, and cytokines in rat NK cells. J Immunol 2005; 174: 7618-7624
  • 95 Boivin DB, James FO, Wu A et al. Circadian clock genes oscillate in human peripheral blood mononuclear cells. Blood 2003; 102: 4143-4145
  • 96 Cutolo M, Straub RH. Circadian rhythms in arthritis: hormonal effects on the immune inflammatory reaction. Autoimmun Rev 2008; 7: 223-228
  • 97 Liu J, Malkani G, Shi X et al. The circadian clock Period 2 gene regulates gamma interferon production of NK cells in host response to lipopolysaccharide-induced endotoxic shock. Infect Immun 2006; 74: 4750-4756
  • 98 Yamada R, Ueda HR. Microarrays: statistical methods for circadian rhythms. Methods Mol Biol 2007; 362: 245-264
  • 99 Spellman PT, Sherlock G, Zhang MQ et al. Comprehensive identification of cell cycle-regulated genes of the yeast Saccharomyces cerevisiae by microarray hybridization. Mol Biol Cell 1998; 9: 3273-3297
  • 100 Ramoni MF, Sebastiani P, Kohane IS. Cluster analysis of gene expression dynamics. Proc Natl Acad Sci USA 2002; 99: 9121-9126
  • 101 Langmead CJ, Yan AK, McClung CR et al. Phase-independent rhythmic analysis of genome-wide expression patterns. J Comput Biol 2003; 10: 521-536
  • 102 Refinetti R, Cornelissen G, Halberg F. Procedures for numerical analysis of circadian rhythms. Biological Rhythm Research 2007; 38: 275-325
  • 103 Yang EH, Almon RR, DuBois DC et al. Identification of global transcriptional dynamics. PLoS ONE 2009; 4: e5992
  • 104 Wang Y, Xu M, Wang Z et al. How to cluster gene expression dynamics in response to environmental signals. Brief Bioinform 2012; 13: 162-174
  • 105 Kim JK, Forger DB. A mechanism for robust circadian timekeeping via stoichiometric balance. Mol Syst Biol 2012; 8: 630
  • 106 Hughes ME, Hogenesch JB, Kornacker K. JTK_CYCLE: an efficient non-parametric algorithm for detecting rhythmic components in genome-scale datasets. J Biol Rhythms 2010; 25: 372-380