51
|
Takeda N, Maemura K. The role of clock genes and circadian rhythm in the development of cardiovascular diseases. Cell Mol Life Sci 2015; 72:3225-34. [PMID: 25972277 PMCID: PMC11113935 DOI: 10.1007/s00018-015-1923-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 05/04/2015] [Accepted: 05/04/2015] [Indexed: 10/23/2022]
Abstract
The time of onset of cardiovascular disorders such as myocardial infarctions or ventricular arrhythmias exhibits a circadian rhythm. Diurnal variations in autonomic nervous activity, plasma cortisol level or renin-angiotensin activity underlie the pathogenesis of cardiovascular diseases. Transcriptional-translational feedback loop of the clock genes constitute a molecular clock system. In addition to the central clock in the suprachiasmatic nucleus, clock genes are also expressed in a circadian fashion in each organ to make up the peripheral clock. The peripheral clock seems to be beneficial for anticipating external stimuli and thus contributes to the maintenance of organ homeostasis. Loss of synchronization between the central and peripheral clocks also augments disease progression. Moreover, accumulating evidence shows that clock genes affect inflammatory and intracellular metabolic signaling. Elucidating the roles of the molecular clock in cardiovascular pathology through the identification of clock controlled genes will help to establish a novel therapeutic approach for cardiovascular disorders.
Collapse
Affiliation(s)
- Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Koji Maemura
- Department of Cardiovascular Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| |
Collapse
|
52
|
Hypertension and Sleep Apnea. Can J Cardiol 2015; 31:889-97. [DOI: 10.1016/j.cjca.2015.05.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 12/28/2022] Open
|
53
|
Alibhai FJ, Tsimakouridze EV, Reitz CJ, Pyle WG, Martino TA. Consequences of Circadian and Sleep Disturbances for the Cardiovascular System. Can J Cardiol 2015; 31:860-72. [DOI: 10.1016/j.cjca.2015.01.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/25/2014] [Accepted: 01/08/2015] [Indexed: 12/01/2022] Open
|
54
|
Schroder EA, Burgess DE, Zhang X, Lefta M, Smith JL, Patwardhan A, Bartos DC, Elayi CS, Esser KA, Delisle BP. The cardiomyocyte molecular clock regulates the circadian expression of Kcnh2 and contributes to ventricular repolarization. Heart Rhythm 2015; 12:1306-14. [PMID: 25701773 PMCID: PMC4541807 DOI: 10.1016/j.hrthm.2015.02.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Sudden cardiac death (SCD) follows a diurnal variation. Data suggest the timing of SCD is influenced by circadian (~24-hour) changes in neurohumoral and cardiomyocyte-specific regulation of the heart's electrical properties. The basic helix-loop-helix transcription factors brain muscle arnt-like1 (BMAL1) and circadian locomotor output control kaput (CLOCK) coordinate the circadian expression of select genes. OBJECTIVE We sought to test whether Bmal1 expression in cardiomyocytes contributes to K(+) channel expression and diurnal changes in ventricular repolarization. METHODS We used transgenic mice that allow for the inducible cardiomyocyte-specific deletion of Bmal1 (iCSΔBmal1(-/-)). We used quantitative polymerase chain reaction, voltage clamping, promoter-reporter bioluminescence assays, and electrocardiographic telemetry. RESULTS Although several K(+) channel gene transcripts were downregulated in iCSΔBmal1(-/-)mouse hearts, only Kcnh2 exhibited a robust circadian pattern of expression that was disrupted in iCSΔBmal1(-/-) hearts. Kcnh2 underlies the rapidly activating delayed-rectifier K(+) current, and the rapidly activating delayed-rectifier K(+) current recorded from iCSΔBmal1(-/-) ventricular cardiomyocytes was ~50% smaller than control ventricular myocytes. Promoter-reporter assays demonstrated that the human Kcnh2 promoter is transactivated by the coexpression of BMAL1 and CLOCK. Electrocardiographic analysis showed that iCSΔBmal1(-/-) mice developed a prolongation in the heart rate-corrected QT interval during the light (resting) phase. This was secondary to an augmented circadian rhythm in the uncorrected QT interval without a corresponding change in the RR interval. CONCLUSION The molecular clock in the heart regulates the circadian expression of Kcnh2, modifies K(+) channel gene expression, and is important for normal ventricular repolarization. Disruption of the cardiomyocyte circadian clock mechanism likely unmasks diurnal changes in ventricular repolarization that could contribute to an increased risk of cardiac arrhythmias/SCD.
Collapse
Affiliation(s)
| | | | - Xiping Zhang
- Center for Muscle Biology, Department of Physiology
| | | | | | | | | | - Claude S Elayi
- Department of Cardiology, University of Kentucky, Lexington, Kentucky
| | | | | |
Collapse
|
55
|
Chen L, Yang G. Recent advances in circadian rhythms in cardiovascular system. Front Pharmacol 2015; 6:71. [PMID: 25883568 PMCID: PMC4381645 DOI: 10.3389/fphar.2015.00071] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/16/2015] [Indexed: 12/20/2022] Open
Abstract
Growing evidence shows that intrinsic circadian clocks are tightly related to cardiovascular functions. The diurnal changes in blood pressure and heart rate are well known circadian rhythms. Endothelial function, platelet aggregation and thrombus formation exhibit circadian changes as well. The onset of many cardiovascular diseases (CVDs) or events, such as myocardial infarction, stroke, arrhythmia, and sudden cardiac death, also exhibits temporal trends. Furthermore, there is strong evidence from animal models and epidemiological studies showing that disruption of circadian rhythms is a significant risk factor for many CVDs, and the intervention of CVDs may have a time dependent effect. In this mini review, we summarized recent advances in our understanding of the relationship between circadian rhythm and cardiovascular physiology and diseases including blood pressure regulation and myocardial infarction.
Collapse
Affiliation(s)
- Lihong Chen
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA ; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA
| | - Guangrui Yang
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA ; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA
| |
Collapse
|
56
|
Martino TA, Young ME. Influence of the Cardiomyocyte Circadian Clock on Cardiac Physiology and Pathophysiology. J Biol Rhythms 2015; 30:183-205. [DOI: 10.1177/0748730415575246] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cardiac function and dysfunction exhibit striking time-of-day-dependent oscillations. Disturbances in both daily rhythms and sleep are associated with increased risk of heart disease, adverse cardiovascular events, and worsening outcomes. For example, the importance of maintaining normal daily rhythms is highlighted by epidemiologic observations that night shift workers present with increased incidence of cardiovascular disease. Rhythmicity in cardiac processes is mediated by a complex interaction between extracardiac (e.g., behaviors and associated neural and humoral fluctuations) and intracardiac influences. Over the course of the day, the intrinsic properties of the myocardium vary at the levels of gene and protein expression, metabolism, responsiveness to extracellular stimuli/stresses, and ion homeostasis, all of which affect contractility (e.g., heart rate and force generation). Over the past decade, the circadian clock within the cardiomyocyte has emerged as an essential mechanism responsible for modulating the intrinsic properties of the heart. Moreover, the critical role of this mechanism is underscored by reports that disruption, through genetic manipulation, results in development of cardiac disease and premature mortality in mice. These findings, in combination with reports that numerous cardiovascular risk factors (e.g., diet, diabetes, aging) distinctly affect the clock in the heart, have led to the hypothesis that aberrant regulation of this mechanism contributes to the etiology of cardiac dysfunction and disease. Here, we provide a comprehensive review on current knowledge regarding known roles of the heart clock and discuss the potential for using these insights for the future development of innovative strategies for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Tami A. Martino
- Cardiovascular Research Group, Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Martin E. Young
- Division of Cardiovascular Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
57
|
HERICHOVÁ I, AMBRUŠOVÁ J, MOLČAN Ľ, VESELÁ A, SVITOK P, ZEMAN M. Different Effects of Phase Advance and Delay in Rotating Light-Dark Regimens on Clock and Natriuretic Peptide Gene Expression in the Rat Heart. Physiol Res 2014; 63:S573-84. [DOI: 10.33549/physiolres.932937] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Under physiological conditions the mammalian circadian system is synchronized to a cyclic environment. The central oscillator in the suprachiasmatic nuclei (SCN) responds predominantly to an external light (L) dark (D) cycle. Peripheral oscillators are more efficiently synchronized by metabolic cues. When the circadian system is exposed to opposing synchronizing cues, peripheral oscillators uncouple from the SCN. To consider influence of phase advances and delays in light regimens mimicking shift work, we analyzed the expression of clock genes (per2, bmal1) and natriuretic peptides (anp, bnp) in the heart of male rats. Experimental groups were exposed to a rotating LD regimen with either 8 h phase advance or delay for 11 weeks. Samples were taken for a 24 h cycle in 4 h intervals. Peripheral oscillators responded to rotating phase advance by decreasing rhythm robustness, while phase delay mostly influenced the phase angle between the acrophase of rhythmic gene expression and the external LD cycle. The expression of anp was arrhythmic in the heart of control rats and was not influenced by rotating LD regimens. The expression of bnp showed a daily rhythm with a nadir during the active phase. The daily rhythm in bnp expression diminished under rotating LD regimen conditions.
Collapse
Affiliation(s)
- I. HERICHOVÁ
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovak Republic
| | | | | | | | | | | |
Collapse
|
58
|
Lecarpentier Y, Claes V, Duthoit G, Hébert JL. Circadian rhythms, Wnt/beta-catenin pathway and PPAR alpha/gamma profiles in diseases with primary or secondary cardiac dysfunction. Front Physiol 2014; 5:429. [PMID: 25414671 PMCID: PMC4220097 DOI: 10.3389/fphys.2014.00429] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/15/2014] [Indexed: 12/13/2022] Open
Abstract
Circadian clock mechanisms are far-from-equilibrium dissipative structures. Peroxisome proliferator-activated receptors (PPAR alpha, beta/delta, and gamma) play a key role in metabolic regulatory processes, particularly in heart muscle. Links between circadian rhythms (CRs) and PPARs have been established. Mammalian CRs involve at least two critical transcription factors, CLOCK and BMAL1 (Gekakis et al., 1998; Hogenesch et al., 1998). PPAR gamma plays a major role in both glucose and lipid metabolisms and presents circadian properties which coordinate the interplay between metabolism and CRs. PPAR gamma is a major component of the vascular clock. Vascular PPAR gamma is a peripheral regulator of cardiovascular rhythms controlling circadian variations in blood pressure and heart rate through BMAL1. We focused our review on diseases with abnormalities of CRs and with primary or secondary cardiac dysfunction. Moreover, these diseases presented changes in the Wnt/beta-catenin pathway and PPARs, according to two opposed profiles. Profile 1 was defined as follows: inactivation of the Wnt/beta-catenin pathway with increased expression of PPAR gamma. Profile 2 was defined as follows: activation of the Wnt/beta-catenin pathway with decreased expression of PPAR gamma. A typical profile 1 disease is arrhythmogenic right ventricular cardiomyopathy, a genetic cardiac disease which presents mutations of the desmosomal proteins and is mainly characterized by fatty acid accumulation in adult cardiomyocytes mainly in the right ventricle. The link between PPAR gamma dysfunction and desmosomal genetic mutations occurs via inactivation of the Wnt/beta-catenin pathway presenting oscillatory properties. A typical profile 2 disease is type 2 diabetes, with activation of the Wnt/beta-catenin pathway and decreased expression of PPAR gamma. CRs abnormalities are present in numerous pathologies such as cardiovascular diseases, sympathetic/parasympathetic dysfunction, hypertension, diabetes, neurodegenerative diseases, cancer which are often closely inter-related.
Collapse
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Centre Hospitalier Régional de Meaux Meaux, France
| | - Victor Claes
- Department of Pharmaceutical Sciences, University of Antwerp Wilrijk, Belgium
| | - Guillaume Duthoit
- Institut de Cardiologie, Hôpital de la Pitié-Salpêtière Paris, France
| | - Jean-Louis Hébert
- Institut de Cardiologie, Hôpital de la Pitié-Salpêtière Paris, France
| |
Collapse
|
59
|
Virag JAI, Lust RM. Circadian influences on myocardial infarction. Front Physiol 2014; 5:422. [PMID: 25400588 PMCID: PMC4214187 DOI: 10.3389/fphys.2014.00422] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/12/2014] [Indexed: 11/13/2022] Open
Abstract
Components of circadian rhythm maintenance, or "clock genes," are endogenous entrainable oscillations of about 24 h that regulate biological processes and are found in the suprachaismatic nucleus (SCN) and many peripheral tissues, including the heart. They are influenced by external cues, or Zeitgebers, such as light and heat, and can influence such diverse phenomena as cytokine expression immune cells, metabolic activity of cardiac myocytes, and vasodilator regulation by vascular endothelial cells. While it is known that the central master clock in the SCN synchronizes peripheral physiologic rhythms, the mechanisms by which the information is transmitted are complex and may include hormonal, metabolic, and neuronal inputs. Whether circadian patterns are causally related to the observed periodicity of events, or whether they are simply epi-phenomena is not well established, but a few studies suggest that the circadian effects likely are real in their impact on myocardial infarct incidence. Cycle disturbances may be harbingers of predisposition and subsequent response to acute and chronic cardiac injury, and identifying the complex interactions of circadian rhythms and myocardial infarction may provide insights into possible preventative and therapeutic strategies for susceptible populations.
Collapse
Affiliation(s)
- Jitka A I Virag
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Robert M Lust
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| |
Collapse
|
60
|
Podobed P, Pyle WG, Ackloo S, Alibhai FJ, Tsimakouridze EV, Ratcliffe WF, Mackay A, Simpson J, Wright DC, Kirby GM, Young ME, Martino TA. The day/night proteome in the murine heart. Am J Physiol Regul Integr Comp Physiol 2014; 307:R121-37. [PMID: 24789993 DOI: 10.1152/ajpregu.00011.2014] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Circadian rhythms are essential to cardiovascular health and disease. Temporal coordination of cardiac structure and function has focused primarily at the physiological and gene expression levels, but these analyses are invariably incomplete, not the least because proteins underlie many biological processes. The purpose of this study was to reveal the diurnal cardiac proteome and important contributions to cardiac function. The 24-h day-night murine cardiac proteome was assessed by two-dimensional difference in gel electrophoresis (2D-DIGE) and liquid chromatography-mass spectrometry. Daily variation was considerable, as ∼7.8% (90/1,147) of spots exhibited statistical changes at paired times across the 24-h light- (L) dark (D) cycle. JTK_CYCLE was used to investigate underlying diurnal rhythms in corresponding mRNA. We next revealed that disruption of the L:D cycle altered protein profiles and diurnal variation in cardiac function in Langendorff-perfused hearts, relative to the L:D cycle. To investigate the role of the circadian clock mechanism, we used cardiomyocyte clock mutant (CCM) mice. CCM myofilaments exhibited a loss of time-of-day-dependent maximal calcium-dependent ATP consumption, and altered phosphorylation rhythms. Moreover, the cardiac proteome was significantly altered in CCM hearts, especially enzymes regulating vital metabolic pathways. Lastly, we used a model of pressure overload cardiac hypertrophy to demonstrate the temporal proteome during heart disease. Our studies demonstrate that time of day plays a direct role in cardiac protein abundance and indicate a novel mechanistic contribution of circadian biology to cardiovascular structure and function.
Collapse
|
61
|
Circadian regulation of myocardial sarcomeric Titin-cap (Tcap, telethonin): identification of cardiac clock-controlled genes using open access bioinformatics data. PLoS One 2014; 9:e104907. [PMID: 25121604 PMCID: PMC4133362 DOI: 10.1371/journal.pone.0104907] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 07/17/2014] [Indexed: 12/26/2022] Open
Abstract
Circadian rhythms are important for healthy cardiovascular physiology and are regulated at the molecular level by a circadian clock mechanism. We and others previously demonstrated that 9–13% of the cardiac transcriptome is rhythmic over 24 h daily cycles; the heart is genetically a different organ day versus night. However, which rhythmic mRNAs are regulated by the circadian mechanism is not known. Here, we used open access bioinformatics databases to identify 94 transcripts with expression profiles characteristic of CLOCK and BMAL1 targeted genes, using the CircaDB website and JTK_Cycle. Moreover, 22 were highly expressed in the heart as determined by the BioGPS website. Furthermore, 5 heart-enriched genes had human/mouse conserved CLOCK:BMAL1 promoter binding sites (E-boxes), as determined by UCSC table browser, circadian mammalian promoter/enhancer database PEDB, and the European Bioinformatics Institute alignment tool (EMBOSS). Lastly, we validated findings by demonstrating that Titin cap (Tcap, telethonin) was targeted by transcriptional activators CLOCK and BMAL1 by showing 1) Tcap mRNA and TCAP protein had a diurnal rhythm in murine heart; 2) cardiac Tcap mRNA was rhythmic in animals kept in constant darkness; 3) Tcap and control Per2 mRNA expression and cyclic amplitude were blunted in ClockΔ19/Δ19 hearts; 4) BMAL1 bound to the Tcap promoter by ChIP assay; 5) BMAL1 bound to Tcap promoter E-boxes by biotinylated oligonucleotide assay; and 6) CLOCK and BMAL1 induced tcap expression by luciferase reporter assay. Thus this study identifies circadian regulated genes in silico, with validation of Tcap, a critical regulator of cardiac Z-disc sarcomeric structure and function.
Collapse
|
62
|
Cleal JK, Shepherd JN, Shearer JL, Bruce KD, Cagampang FR. Sensitivity of housekeeping genes in the suprachiasmatic nucleus of the mouse brain to diet and the daily light-dark cycle. Brain Res 2014; 1575:72-7. [PMID: 24881883 DOI: 10.1016/j.brainres.2014.05.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/20/2014] [Accepted: 05/21/2014] [Indexed: 02/07/2023]
Abstract
The endogenous timing system within the suprachiasmatic nuclei (SCN) of the hypothalamus drives the cyclic expression of the clock molecules across the 24h day-night cycle controlling downstream molecular pathways and physiological processes. The developing fetal clock system is sensitive to the environment and physiology of the pregnant mother and as such disruption of this system could lead to altered physiology in the offspring. Characterizing the gene profiles of the endogenous molecular clock system by quantitative reverse transcription polymerase chain reaction is dependent on normalization by appropriate housekeeping genes (HKGs). However, many HKGs commonly used as internal controls, although stably expressed under control conditions, can vary significantly in their expression under certain experimental conditions. Here we analyzed the expression of 10 classic HKG across the 24h light-dark cycle in the SCN of mouse offspring exposed to normal chow or a high fat diet during early development and in postnatal life. We found that the HKGs glyceraldehyde-3-phosphate dehydrogenase, beta actin and adenosine triphosphate synthase subunit to be the most stably expressed genes in the SCN regardless of diet or time within the 24h light-dark cycle, and are therefore suitable to be used as internal controls. However SCN samples collected during the light and dark periods did show differences in expression and as such the timing of collection should be considered when carrying out gene expression studies.
Collapse
Affiliation(s)
- Jane K Cleal
- Institute of Developmental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital (mailpoint 887), Southampton SO16 6YD, UK.
| | - James N Shepherd
- Institute of Developmental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital (mailpoint 887), Southampton SO16 6YD, UK
| | - Jasmine L Shearer
- Institute of Developmental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital (mailpoint 887), Southampton SO16 6YD, UK
| | - Kimberley D Bruce
- Institute of Developmental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital (mailpoint 887), Southampton SO16 6YD, UK
| | - Felino R Cagampang
- Institute of Developmental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital (mailpoint 887), Southampton SO16 6YD, UK
| |
Collapse
|
63
|
Carver KA, Lourim D, Tryba AK, Harder DR. Rhythmic expression of cytochrome P450 epoxygenases CYP4x1 and CYP2c11 in the rat brain and vasculature. Am J Physiol Cell Physiol 2014; 307:C989-98. [PMID: 25055826 DOI: 10.1152/ajpcell.00401.2013] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Mammals have circadian variation in blood pressure, heart rate, vascular tone, thrombotic tendency, and cerebral blood flow (CBF). These changes may be in part orchestrated by circadian variation in clock gene expression within cells comprising the vasculature that modulate blood flow (e.g., fibroblasts, cerebral vascular smooth muscle cells, astrocytes, and endothelial cells). However, the downstream mechanisms that underlie circadian changes in blood flow are unknown. Cytochrome P450 epoxygenases (Cyp4x1 and Cyp2c11) are expressed in the brain and vasculature and metabolize arachidonic acid (AA) to form epoxyeicosatrienoic acids (EETs). EETs are released from astrocytes, neurons, and vascular endothelial cells and act as potent vasodilators, increasing blood flow. EETs released in response to increases in neural activity evoke a corresponding increase in blood flow known as the functional hyperemic response. We examine the hypothesis that Cyp2c11 and Cyp4x1 expression and EETs production vary in a circadian manner in the rat brain and cerebral vasculature. RT-PCR revealed circadian/diurnal expression of clock and clock-controlled genes as well as Cyp4x1 and Cyp2c11, within the rat hippocampus, middle cerebral artery, inferior vena cava, hippocampal astrocytes and rat brain microvascular endothelial cells. Astrocyte and endothelial cell culture experiments revealed rhythmic variation in Cyp4x1 and Cyp2c11 gene and protein expression with a 12-h period and parallel rhythmic production of EETs. Our data suggest there is circadian regulation of Cyp4x1 and Cyp2c11 gene expression. Such rhythmic EETs production may contribute to circadian changes in blood flow and alter risk of adverse cardiovascular events throughout the day.
Collapse
Affiliation(s)
- Koryn A Carver
- Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - David Lourim
- Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - Andrew K Tryba
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and Clement J. Zablocki VA Medical Center, Milwaukee, Wisconsin
| | - David R Harder
- Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and Clement J. Zablocki VA Medical Center, Milwaukee, Wisconsin
| |
Collapse
|
64
|
Kurbatova IV, Topchieva LV, Korneva VA, Kolomeichuk SN, Nemova NN. Expression of circadian rhythm genes CLOCK, BMAL1, and PER1 in buccal epithelial cells of patients with essential arterial hypertension in dependence on polymorphic variants of CLOCK and BMAL1 genes. Bull Exp Biol Med 2014; 157:360-3. [PMID: 25070164 DOI: 10.1007/s10517-014-2566-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Indexed: 11/30/2022]
Abstract
The transcript levels of circadian rhythm genes CLOCK, BMAL1, and PER1 in buccal epithelial cells of the patients with essential arterial hypertension was analyzed in relation to polymorphic variants of CLOCK and BMAL1 genes. These levels were assessed with realtime PCR method at daily hours 9, 13, and 17. The significant differences were revealed in transcript levels of the examined genes in patients with various genotypes at the polymorphic markers 3111TC and 257TG regulatory regions of CLOCK gene. The study detected no significant differences among the carriers of various genotypes at polymorphic markers 862TC and 2121GA of CLOCK gene and 56445TC of BMAL1 gene.
Collapse
Affiliation(s)
- I V Kurbatova
- Institute of Biology, Karelian Scientific Center, Russian Academy of Sciences, Petrozavodsk, Russia,
| | | | | | | | | |
Collapse
|
65
|
Lagishetty V, Parthasarathy PT, Phillips O, Fukumoto J, Cho Y, Fukumoto I, Bao H, Cox R, Galam L, Lockey RF, Kolliputi N. Dysregulation of CLOCK gene expression in hyperoxia-induced lung injury. Am J Physiol Cell Physiol 2014; 306:C999-C1007. [PMID: 24696144 DOI: 10.1152/ajpcell.00064.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyperoxic acute lung injury (HALI) is characterized by inflammation and epithelial cell death. CLOCK genes are master regulators of circadian rhythm also implicated in inflammation and lung diseases. However, the relationship of CLOCK genes in hyperoxia-induced lung injury has not been studied. This study will determine if HALI alters CLOCK gene expression. To test this, wild-type and NALP3(-/-) mice were exposed to room air or hyperoxia for 24, 48, or 72 h. In addition, mice were exposed to different concentrations of hyperoxia (50, 75, or 100% O2) or room air for 72 h. The mRNA and protein levels of lung CLOCK genes, based on quantitative PCR and Western blot analysis, respectively, and their target genes are significantly elevated in mice exposed to hyperoxia compared with controls. Alterations in CLOCK genes are associated with increased inflammatory markers in bronchoalveolar lavage fluid of hyperoxic mice compared with controls. Histological examination of mice lungs exposed to hyperoxia show increased inflammation and alveolar congestion compared with controls. Our results indicate sequential increase in CLOCK gene expression in lungs of mice exposed to hyperoxia compared with controls. Additionally, data suggest a dose-dependent increase in CLOCK gene expression with increased oxygen concentrations. To validate if the expression changes related to CLOCK genes are indeed associated with inflammation, NALP3(-/-) was introduced to analyze loss of function in inflammation. Western blot analysis showed significant CLOCK gene downregulation in NALP3(-/-) mice compared with wild-type controls. Together, our results demonstrate that hyperoxia-mediated lung inflammation is associated with alterations in CLOCK gene expression.
Collapse
Affiliation(s)
- Venu Lagishetty
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Prasanna Tamarapu Parthasarathy
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Oluwakemi Phillips
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Jutaro Fukumoto
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Young Cho
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Itsuko Fukumoto
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Huynh Bao
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Ruan Cox
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Lakshmi Galam
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Richard F Lockey
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
66
|
Lipkova J, Bienertova-Vasku JA, Spinarova L, Bienert P, Hlavna M, Pavkova Goldbergova M, Parenica J, Spinar J, Vasku A. Per3 VNTR polymorphism and chronic heart failure. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2014; 158:80-3. [DOI: 10.5507/bp.2012.069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 07/04/2012] [Indexed: 01/03/2023] Open
|
67
|
Yang Y, Cheng S, Li Y, Jiang Z, Liu Y, Wang Y, Xiao J, Guo H, Wang Z. The influence of L-arginine on circadian rhythm and circadian periodgenes. BIOL RHYTHM RES 2013. [DOI: 10.1080/09291016.2012.704799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
68
|
Herichová I, Šoltésová D, Szántóová K, Mravec B, Neupauerová D, Veselá A, Zeman M. Effect of angiotensin II on rhythmic per2 expression in the suprachiasmatic nucleus and heart and daily rhythm of activity in Wistar rats. ACTA ACUST UNITED AC 2013; 186:49-56. [PMID: 23850797 DOI: 10.1016/j.regpep.2013.06.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 04/29/2013] [Accepted: 06/27/2013] [Indexed: 11/26/2022]
Abstract
Endogenous daily rhythms are generated by the hierarchically organized circadian system predominantly synchronized by the external light (L): dark (D) cycle. During recent years several humoral signals have been found to influence the generation and manifestation of daily rhythm. Since most studies have been performed under in vitro conditions, the mechanisms employed under in vivo conditions need to be investigated. Our study focused on angiotensin II (angII)-mediated regulation of Per2 expression in the suprachiasmatic nuclei (SCN) and heart and spontaneous locomotor activity in Wistar rats under synchronized conditions. Angiotensin II was infused (100ng/kg/min) via subcutaneously implanted osmotic minipumps for 7 or 28days. Samples were taken in 4-h intervals during a 24hcycle and after a light pulse applied in the first and second part of the dark phase. Gene expression was measured using real time PCR. Locomotor activity was monitored using an infrared camera with a remote control installed in the animal facility. Seven days of angII infusion caused an increase in blood pressure and heart/body weight index and 28days of angII infusion also increased water intake in comparison with controls. We observed a distinct daily rhythm in Per2 expression in the SCN and heart of control rats and infused rats. Seven days of angII infusion did not influence Per2 expression in the heart. 28days of angII treatment caused significant phase advance and a decrease in nighttime expression of Per2 and influenced expression of clock controlled genes Rev-erb alpha and Dbp in the heart compared to the control. Four weeks of angII infusion decreased the responsiveness of Per2 expression in the SCN to a light pulse at the end of the dark phase of the 24hcycle. Expression of mRNA coding angiotensin-converting enzyme (ACE) and angiotensin-converting enzyme 2 (ACE2) showed a daily rhythm in the heart of control rats. Four weeks of angII infusion caused a decrease in amplitude of rhythmic expression of Ace, the disappearance of rhythm and an increase in Ace2 expression. The Ace/Ace2 ratio showed a rhythmic pattern in the heart of control rats with peak levels during the dark phase. Angiotensin II infusion decreased the mean Ace/Ace2 mRNA ratio in the heart. We observed a significant daily rhythm in expression of brain natriuretic peptide (BNP) in the heart of control rats. In hypertensive rats mean value of Bnp expression increased. Locomotor activity showed a distinct daily rhythm in both groups. Angiotensin II time dependently decreased ratio of locomotor activity in active versus passive phase of 24hcycle. To conclude, 28days of subcutaneous infusion of angII modulates the functioning of the central and peripheral circadian system measured at the level of Per2 expression and locomotor activity.
Collapse
Affiliation(s)
- Iveta Herichová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Mlynská dolina B-2, 842 15 Bratislava, Slovak Republic.
| | | | | | | | | | | | | |
Collapse
|
69
|
Diurnal variation in excitation-contraction coupling is lost in the adult spontaneously hypertensive rat heart. J Hypertens 2013; 31:1214-23. [DOI: 10.1097/hjh.0b013e328360ae4b] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
70
|
Gamble KL, Young ME. Metabolism as an integral cog in the mammalian circadian clockwork. Crit Rev Biochem Mol Biol 2013; 48:317-31. [PMID: 23594144 DOI: 10.3109/10409238.2013.786672] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Circadian rhythms are an integral part of life. These rhythms are apparent in virtually all biological processes studies to date, ranging from the individual cell (e.g. DNA synthesis) to the whole organism (e.g. behaviors such as physical activity). Oscillations in metabolism have been characterized extensively in various organisms, including mammals. These metabolic rhythms often parallel behaviors such as sleep/wake and fasting/feeding cycles that occur on a daily basis. What has become increasingly clear over the past several decades is that many metabolic oscillations are driven by cell-autonomous circadian clocks, which orchestrate metabolic processes in a temporally appropriate manner. During the process of identifying the mechanisms by which clocks influence metabolism, molecular-based studies have revealed that metabolism should be considered an integral circadian clock component. The implications of such an interrelationship include the establishment of a vicious cycle during cardiometabolic disease states, wherein metabolism-induced perturbations in the circadian clock exacerbate metabolic dysfunction. The purpose of this review is therefore to highlight recent insights gained regarding links between cell-autonomous circadian clocks and metabolism and the implications of clock dysfunction in the pathogenesis of cardiometabolic diseases.
Collapse
Affiliation(s)
- Karen L Gamble
- Division of Behavioral Neurobiology, Department of Psychiatry, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
71
|
Shusterman V, Lampert R. Role of Stress in Cardiac Arrhythmias. J Atr Fibrillation 2013; 5:834. [PMID: 28496839 DOI: 10.4022/jafib.834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 03/03/2013] [Accepted: 03/05/2013] [Indexed: 11/10/2022]
Abstract
Stress is a major trigger of cardiac arrhythmias; it exerts profound effects on electrophysiology of the cardiomyocytes and the cardiac rhythm. Psychological and physiological stressors impact the cardiovascular system through the autonomic nervous system (ANS). While stressors vary, properties of the stress response at the level of cardiovascular system (collectively referred to as the autonomic cardiovascular responses) are similar and can be studied independently from the properties of specific stressors. Here, we will review the clinical and experimental evidence linking common stressors and atrial arrhythmias. Specifically, we will describe the impact of psychological and circadian stressors on ANS activity and arrhythmogenesis. We will also review studies examining relationships between autonomic cardiovascular responses and cardiac arrhythmias in ambulatory and laboratory settings.
Collapse
Affiliation(s)
- Vladimir Shusterman
- University of Pittsburgh School of Medicine and Yale University School of Medicine
| | - Rachel Lampert
- University of Pittsburgh School of Medicine and Yale University School of Medicine
| |
Collapse
|
72
|
Hardeland R. Chronobiology of Melatonin beyond the Feedback to the Suprachiasmatic Nucleus-Consequences to Melatonin Dysfunction. Int J Mol Sci 2013; 14:5817-41. [PMID: 23481642 PMCID: PMC3634486 DOI: 10.3390/ijms14035817] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 03/01/2013] [Accepted: 03/04/2013] [Indexed: 12/28/2022] Open
Abstract
The mammalian circadian system is composed of numerous oscillators, which gradually differ with regard to their dependence on the pacemaker, the suprachiasmatic nucleus (SCN). Actions of melatonin on extra-SCN oscillators represent an emerging field. Melatonin receptors are widely expressed in numerous peripheral and central nervous tissues. Therefore, the circadian rhythm of circulating, pineal-derived melatonin can have profound consequences for the temporal organization of almost all organs, without necessarily involving the melatonin feedback to the suprachiasmatic nucleus. Experiments with melatonin-deficient mouse strains, pinealectomized animals and melatonin receptor knockouts, as well as phase-shifting experiments with explants, reveal a chronobiological role of melatonin in various tissues. In addition to directly steering melatonin-regulated gene expression, the pineal hormone is required for the rhythmic expression of circadian oscillator genes in peripheral organs and to enhance the coupling of parallel oscillators within the same tissue. It exerts additional effects by modulating the secretion of other hormones. The importance of melatonin for numerous organs is underlined by the association of various diseases with gene polymorphisms concerning melatonin receptors and the melatonin biosynthetic pathway. The possibilities and limits of melatonergic treatment are discussed with regard to reductions of melatonin during aging and in various diseases.
Collapse
Affiliation(s)
- Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, Berliner Str. 28, Göttingen D-37073, Germany.
| |
Collapse
|
73
|
Chatham JC, Young ME. Regulation of myocardial metabolism by the cardiomyocyte circadian clock. J Mol Cell Cardiol 2013; 55:139-46. [PMID: 22766272 PMCID: PMC4107417 DOI: 10.1016/j.yjmcc.2012.06.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 06/19/2012] [Accepted: 06/20/2012] [Indexed: 11/24/2022]
Abstract
On a daily basis, the heart is subjected to dramatic fluctuations in energetic demand and neurohumoral influences, many of which occur in a temporally predictable manner. In order to preserve cardiac performance, the heart must therefore maintain metabolic flexibility, even within the confines of a single day. Recent studies have established mechanistic links between time-of-day-dependent oscillations in myocardial metabolism and the cardiomyocyte circadian clock. More specifically, evidence suggests that this cell autonomous molecular mechanism regulates myocardial glucose uptake, flux through both glycolysis and the hexosamine biosynthetic pathway, and pyruvate oxidation, as well as glycogen, triglyceride, and protein turnover. These observations have led to the hypothesis that the cardiomyocyte circadian clock confers the selective advantage of anticipation of increased energetic demand during the awake period. Here, we review the accumulative evidence in support of this hypothesis thus far, and discuss the possibility that attenuation of these metabolic rhythms, through disruption of the cardiomyocyte circadian clock, contributes towards the etiology of cardiac dysfunction in various disease states. This article is part of a Special Issue entitled "Focus on Cardiac Metabolism".
Collapse
Affiliation(s)
- John C. Chatham
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Martin E. Young
- Division of Cardiovascular Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
74
|
Liu Z, Chu G. Chronobiology in mammalian health. Mol Biol Rep 2012; 40:2491-501. [DOI: 10.1007/s11033-012-2330-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 11/19/2012] [Indexed: 11/30/2022]
|
75
|
Kurbatova IV, Kolomeichuk SN, Topchieva LV, Korneva VA, Nemova NN. Expression of the CLOCK, BMAL1, and PER1 circadian genes in human oral mucosa cells as dependent on CLOCK gene polymorphic variants. DOKLADY BIOLOGICAL SCIENCES : PROCEEDINGS OF THE ACADEMY OF SCIENCES OF THE USSR, BIOLOGICAL SCIENCES SECTIONS 2012; 446:323-6. [PMID: 23129285 DOI: 10.1134/s0012496612050146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Indexed: 11/23/2022]
Affiliation(s)
- I V Kurbatova
- Karelian Scientific Center, Russian Academy of Sciences, Petrozavodsk, Russia
| | | | | | | | | |
Collapse
|
76
|
Bray MS, Ratcliffe WF, Grenett MH, Brewer RA, Gamble KL, Young ME. Quantitative analysis of light-phase restricted feeding reveals metabolic dyssynchrony in mice. Int J Obes (Lond) 2012; 37:843-52. [PMID: 22907695 PMCID: PMC3505273 DOI: 10.1038/ijo.2012.137] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Background Considerable evidence suggests that the time of day at which calories are consumed markedly impacts body weight gain and adiposity. However, a precise quantification of energy balance parameters during controlled animal studies enforcing time-of-day-restricted feeding is currently lacking in the absence of direct human interaction. Objective The purpose of the present study was therefore to quantify the effects of restricted feeding during the light (sleep) phase in a fully-automated, computer-controlled comprehensive laboratory animal monitoring system (CLAMS) designed to modulate food access in a time-of-day-dependent manner. Energy balance, gene expression (within metabolically-relevant tissues), humoral factors, and body weight were assessed. Results We report that relative to mice fed only during the dark (active) phase, light (sleep) phase fed mice: 1) consume a large meal upon initiation of food availability; 2) consume greater total calories per day; 3) exhibit a higher RER (indicative of decreased reliance on lipid/fatty acid oxidation); 4) exhibit tissue-specific alterations in the phases and amplitudes of circadian clock and metabolic genes in metabolically active tissues (greatest phase differences observed in the liver, and diminution of amplitudes in epididymal fat, gastrocnemius muscle, and heart); 5) exhibit diminished amplitude in humoral factor diurnal variations (e.g., corticosterone); and 6) exhibit greater weight gain within 9 days of restricted feeding. Conclusions Collectively, these data suggest that weight gain following light (sleep) phase restricted feeding is associated with significant alterations in energy balance, as well as dyssynchrony between metabolically active organs.
Collapse
Affiliation(s)
- M S Bray
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
77
|
Tong M, Watanabe E, Yamamoto N, Nagahata-Ishiguro M, Maemura K, Takeda N, Nagai R, Ozaki Y. Circadian expressions of cardiac ion channel genes in mouse might be associated with the central clock in the SCN but not the peripheral clock in the heart. BIOL RHYTHM RES 2012; 44:519-530. [PMID: 23420534 PMCID: PMC3570950 DOI: 10.1080/09291016.2012.704801] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 06/13/2012] [Indexed: 11/03/2022]
Abstract
Significant circadian variations exist in the frequency of cardiac arrhythmia, but few studies have examined the relation between cardiac ion channels genes and biological clocks. We investigated this relation using suprachiasmatic nuclei lesion (SCNX) and pharmacological autonomic nervous system block (ANSB) mice. Significant 24-h variations were observed in the expression of clock genes Per2, Bmal1, and Dbp and ion channel genes KCNA5, KCND2, KCHIP2, and KCNK3 in the control mice hearts. In the SCNX mice, all genes examined lost circadian rhythm. In the ANSB mice, the expressions of the three clock genes were dampened significantly but still had circadian rhythm, whereas the four ion channel gene expressions lost rhythm. Heart rate also lost circadian rhythm in both the SCNX and ANSB mice. These results suggest that some ion channel gene expressions might be regulated by the central clock in the SCN through the ANS but not the peripheral clock in the heart.
Collapse
Affiliation(s)
- Maoqing Tong
- Department of Cardiology, Fujita Health University School of Medicine, Toyoake, Japan
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Tsimakouridze EV, Straume M, Podobed PS, Chin H, LaMarre J, Johnson R, Antenos M, Kirby GM, Mackay A, Huether P, Simpson JA, Sole M, Gadal G, Martino TA. Chronomics of Pressure Overload–Induced Cardiac Hypertrophy in Mice Reveals Altered Day/Night Gene Expression and Biomarkers of Heart Disease. Chronobiol Int 2012; 29:810-21. [DOI: 10.3109/07420528.2012.691145] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
79
|
Abstract
Mammals have an endogenous timing system in the suprachiasmatic nuclei (SCN) of the hypothalamic region of the brain. This internal clock system is composed of an intracellular feedback loop that drives the expression of molecular components and their constitutive protein products to oscillate over a period of about 24 h (hence the term 'circadian'). These circadian oscillations bring about rhythmic changes in downstream molecular pathways and physiological processes such as those involved in nutrition and metabolism. It is now emerging that the molecular components of the clock system are also found within the cells of peripheral tissues, including the gastrointestinal tract, liver and pancreas. The present review examines their role in regulating nutritional and metabolic processes. In turn, metabolic status and feeding cycles are able to feed back onto the circadian clock in the SCN and in peripheral tissues. This feedback mechanism maintains the integrity and temporal coordination between various components of the circadian clock system. Thus, alterations in environmental cues could disrupt normal clock function, which may have profound effects on the health and well-being of an individual.
Collapse
|
80
|
Iimura T, Nakane A, Sugiyama M, Sato H, Makino Y, Watanabe T, Takagi Y, Numano R, Yamaguchi A. A fluorescence spotlight on the clockwork development and metabolism of bone. J Bone Miner Metab 2012; 30:254-69. [PMID: 21766187 DOI: 10.1007/s00774-011-0295-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 06/10/2011] [Indexed: 01/26/2023]
Abstract
Biological phenomena that exhibit periodic activity are often referred as biorhythms or biological clocks. Among these, circadian rhythms, cyclic patterns reflecting a 24-h cycle, are the most obvious in many physiological activities including bone growth and metabolism. In the late 1990s, several clock genes were isolated and their primary structures and functions were identified. The feedback loop model of transcriptional factors was proposed to work as a circadian core oscillator not only in the suprachiasmatic nuclei of the anterior hypothalamus, which is recognized as the mammalian central clock, but also in various peripheral tissues including cartilage and bone. Looking back to embryonic development, the fundamental architecture of skeletal patterning is regulated by ultradian clocks that are defined as biorhythms that cycle more than once every 24 h. As post-genomic approaches, transcriptome analysis by micro-array and bioimaging assays to detect luminescent and fluorescent signals have been exploited to uncover a more comprehensive set of genes and spatio-temporal regulation of the clockwork machinery in animal models. In this review paper, we provide an overview of topics related to these molecular clocks in skeletal biology and medicine, and discuss how fluorescence imaging approaches can contribute to widening our views of this realm of biomedical science.
Collapse
Affiliation(s)
- Tadahiro Iimura
- Global Center of Excellence (GCOE) Program, International Research Center for Molecular Science in Tooth and Bone Diseases, Tokyo Medical and Dental University, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Meermeier N, Krishnan N. Circadian regulation of cellular homeostasis--implications for cell metabolism and clinical diseases. Med Hypotheses 2012; 79:17-24. [PMID: 22521428 DOI: 10.1016/j.mehy.2012.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 03/25/2012] [Indexed: 01/07/2023]
Abstract
The major pathways involving nutrient and energy metabolism including cellular homeostasis are profoundly impacted by the circadian clock, which orchestrates diurnal rhythms in physiology and behavior. While the links between circadian and metabolic rhythms are unclear, recent studies imply a close link between the two with one feeding back on the other. In this discussion, we present the hypothesis that circadian clocks likely contribute to cellular homeostasis, especially proteostasis, through regulation of metabolic rhythms, which in turn feed-back on circadian oscillators. The disruption of circadian clocks leads to altered metabolic rhythms and metabolic disease states as a result of altered cellular homeostasis.
Collapse
Affiliation(s)
- N Meermeier
- Department of Microbiology, 220 Nash Hall, Oregon State University, Corvallis, OR 97331, United States.
| | | |
Collapse
|
82
|
Silver AC, Arjona A, Hughes ME, Nitabach MN, Fikrig E. Circadian expression of clock genes in mouse macrophages, dendritic cells, and B cells. Brain Behav Immun 2012; 26:407-13. [PMID: 22019350 PMCID: PMC3336152 DOI: 10.1016/j.bbi.2011.10.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 09/28/2011] [Accepted: 10/05/2011] [Indexed: 12/22/2022] Open
Abstract
In mammals, circadian and daily rhythms influence nearly all aspects of physiology, ranging from behavior to gene expression. Functional molecular clocks have been described in the murine spleen and splenic NK cells. The aim of our study was to investigate the existence of molecular clock mechanisms in other immune cells. Therefore, we measured the circadian changes in gene expression of clock genes (Per1, Per2, Bmal1, and Clock) and clock-controlled transcription factors (Rev-erbα and Dbp) in splenic enriched macrophages, dendritic cells, and B cells in both mice entrained to a light-dark cycle and under constant environmental conditions. Our study reveals the existence of functional molecular clock mechanisms in splenic macrophages, dendritic cells, and B cells.
Collapse
Affiliation(s)
- Adam C Silver
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8022, USA.
| | | | | | | | | |
Collapse
|
83
|
Kohsaka A, Waki H, Cui H, Gouraud SS, Maeda M. Integration of metabolic and cardiovascular diurnal rhythms by circadian clock. Endocr J 2012; 59:447-56. [PMID: 22361995 DOI: 10.1507/endocrj.ej12-0057] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Understanding how the 24-hour blood-pressure rhythm is programmed has been one of the most challenging questions in cardiovascular research. The 24-hour blood-pressure rhythm is primarily driven by the circadian clock system, in which the master circadian pacemaker within the suprachiasmatic nuclei of the hypothalamus is first entrained to the light/dark cycle and then transmits synchronizing signals to the peripheral clocks common to most tissues, including the heart and blood vessels. However, the circadian system is more complex than this basic hierarchical structure, as indicated by the discovery that peripheral clocks are either influenced to some degree or fully driven by temporal changes in energy homeostasis, independent of the light entrainment pathway. Through various comparative genomic approaches and through studies exploiting mouse genetics and transgenics, we now appreciate that cardiovascular tissues possess a large number of metabolic genes whose expression cycle and reciprocally affect the transcriptional control of major circadian clock genes. These findings indicate that metabolic cycles can directly or indirectly affect the diurnal rhythm of cardiovascular function. Here, we discuss a framework for understanding how the 24-hour blood-pressure rhythm is driven by the circadian system that integrates cardiovascular and metabolic function.
Collapse
Affiliation(s)
- Akira Kohsaka
- Department of Physiology, Wakayama Medical University School of Medicine, Japan.
| | | | | | | | | |
Collapse
|
84
|
Durgan DJ, Pat BM, Laczy B, Bradley JA, Tsai JY, Grenett MH, Ratcliffe WF, Brewer RA, Nagendran J, Villegas-Montoya C, Zou C, Zou L, Johnson RL, Dyck JRB, Bray MS, Gamble KL, Chatham JC, Young ME. O-GlcNAcylation, novel post-translational modification linking myocardial metabolism and cardiomyocyte circadian clock. J Biol Chem 2011; 286:44606-19. [PMID: 22069332 PMCID: PMC3247942 DOI: 10.1074/jbc.m111.278903] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Revised: 10/21/2011] [Indexed: 01/02/2023] Open
Abstract
The cardiomyocyte circadian clock directly regulates multiple myocardial functions in a time-of-day-dependent manner, including gene expression, metabolism, contractility, and ischemic tolerance. These same biological processes are also directly influenced by modification of proteins by monosaccharides of O-linked β-N-acetylglucosamine (O-GlcNAc). Because the circadian clock and protein O-GlcNAcylation have common regulatory roles in the heart, we hypothesized that a relationship exists between the two. We report that total cardiac protein O-GlcNAc levels exhibit a diurnal variation in mouse hearts, peaking during the active/awake phase. Genetic ablation of the circadian clock specifically in cardiomyocytes in vivo abolishes diurnal variations in cardiac O-GlcNAc levels. These time-of-day-dependent variations appear to be mediated by clock-dependent regulation of O-GlcNAc transferase and O-GlcNAcase protein levels, glucose metabolism/uptake, and glutamine synthesis in an NAD-independent manner. We also identify the clock component Bmal1 as an O-GlcNAc-modified protein. Increasing protein O-GlcNAcylation (through pharmacological inhibition of O-GlcNAcase) results in diminished Per2 protein levels, time-of-day-dependent induction of bmal1 gene expression, and phase advances in the suprachiasmatic nucleus clock. Collectively, these data suggest that the cardiomyocyte circadian clock increases protein O-GlcNAcylation in the heart during the active/awake phase through coordinated regulation of the hexosamine biosynthetic pathway and that protein O-GlcNAcylation in turn influences the timing of the circadian clock.
Collapse
Affiliation(s)
- David J. Durgan
- From the Division of Cardiovascular Diseases, Department of Medicine
| | - Betty M. Pat
- From the Division of Cardiovascular Diseases, Department of Medicine
| | - Boglarka Laczy
- the Division of Molecular and Cellular Pathology, Department of Pathology
| | - Jerry A. Bradley
- From the Division of Cardiovascular Diseases, Department of Medicine
| | - Ju-Yun Tsai
- the United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, and
| | | | | | - Rachel A. Brewer
- From the Division of Cardiovascular Diseases, Department of Medicine
| | - Jeevan Nagendran
- the Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Carolina Villegas-Montoya
- the United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, and
| | - Chenhang Zou
- the Division of Molecular and Cellular Pathology, Department of Pathology
| | - Luyun Zou
- the Division of Molecular and Cellular Pathology, Department of Pathology
| | | | - Jason R. B. Dyck
- the Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Molly S. Bray
- the Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Karen L. Gamble
- the Division of Behavioral Neurobiology, Department of Psychiatry, and
| | - John C. Chatham
- the Division of Molecular and Cellular Pathology, Department of Pathology
| | - Martin E. Young
- From the Division of Cardiovascular Diseases, Department of Medicine
| |
Collapse
|
85
|
Su W, Xie Z, Guo Z, Duncan MJ, Lutshumba J, Gong MC. Altered clock gene expression and vascular smooth muscle diurnal contractile variations in type 2 diabetic db/db mice. Am J Physiol Heart Circ Physiol 2011; 302:H621-33. [PMID: 22140039 DOI: 10.1152/ajpheart.00825.2011] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study was designed to determine whether the 24-h rhythms of clock gene expression and vascular smooth muscle (VSM) contractile responses are altered in type 2 diabetic db/db mice. Control and db/db mice were euthanized at 6-h intervals throughout the day. The aorta, mesenteric arteries, heart, kidney, and brain were isolated. Clock and target gene mRNA levels were determined by either real-time PCR or in situ hybridization. Isometric contractions were measured in isolated aortic helical strips, and pressor responses to an intravenous injection of vasoconstrictors were determined in vivo using radiotelemetry. We found that the 24-h mRNA rhythms of the following genes were suppressed in db/db mice compared with control mice: the clock genes period homolog 1/2 (Per1/2) and cryptochrome 1/2 (Cry1/2) and their target genes D site albumin promoter-binding protein (Dbp) and peroxisome proliferator-activated receptor-γ (Pparg) in the aorta and mesenteric arteries; Dbp in the heart; Per1, nuclear receptor subfamily 1, group D, member 1 (Rev-erba), and Dbp in the kidney; and Per1 in the suprachiasmatic nucleus. The 24-h contractile variations in response to phenylephrine (α(1)-agonist), ANG II, and high K(+) were significantly altered in the aortas from db/db mice compared with control mice. The diurnal variations of the in vivo pressor responses to phenylephrine and ANG II were lost in db/db mice. Moreover, the 24-h mRNA rhythms of the contraction-related proteins Rho kinase 1/2, PKC-potentiated phosphatase inhibitory protein of 17 kDa, calponin-3, tropomyosin-1/2, and smooth muscle protein 22-α were suppressed in db/db mice compared with control mice. Together, our data demonstrated that the 24-h rhythms of clock gene mRNA, mRNA levels of several contraction-related proteins, and VSM contraction were disrupted in db/db mice, which may contribute to the disruption of their blood pressure circadian rhythm.
Collapse
Affiliation(s)
- Wen Su
- Departments of Physiology, University of Kentucky Medical Center, Lexington, KY 40536, USA
| | | | | | | | | | | |
Collapse
|
86
|
Bae JH, Ahn K, Nam GH, Lee CE, Park KD, Lee HK, Cho BW, Kim HS. Molecular characterization of alternative transcripts of the horse BMAL1 gene. Zoolog Sci 2011; 28:671-5. [PMID: 21882956 DOI: 10.2108/zsj.28.671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The horse BMAL1 gene encodes the brain and muscle Arnt-like protein 1, which is a key regulator of circadian rhythmic systems in most organs and cells. The first exon of the horse-specific BMAL1 gene is produced by an exonization event of LINE3 (CR1) and SINE (MIR) was detected by bioinformatic analysis. Alternative variants generated by cassette exon event in various horse tissues were also detected by RT-PCR amplification and sequencing. The cDNA sequences of the horse transcripts (BMAL1a, BMAL1b) contain additional 21 bp and 71 bp fragments relative to horse BMAL1. Quantitative real-time RT-PCR was performed to compare the expression patterns between transcript variants in various horse tissues. The results of these experiments showed splice variants that were widely expressed in most tissues. Furthermore, they were highly expressed in cerebellum, heart, and kidney.
Collapse
Affiliation(s)
- Jin-Han Bae
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 609-735, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Durgan DJ, Tsai JY, Grenett MH, Pat BM, Ratcliffe WF, Villegas-Montoya C, Garvey ME, Nagendran J, Dyck JRB, Bray MS, Gamble KL, Gimble JM, Young ME. Evidence suggesting that the cardiomyocyte circadian clock modulates responsiveness of the heart to hypertrophic stimuli in mice. Chronobiol Int 2011; 28:187-203. [PMID: 21452915 DOI: 10.3109/07420528.2010.550406] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Circadian dyssynchrony of an organism (at the whole-body level) with its environment, either through light-dark (LD) cycle or genetic manipulation of clock genes, augments various cardiometabolic diseases. The cardiomyocyte circadian clock has recently been shown to influence multiple myocardial processes, ranging from transcriptional regulation and energy metabolism to contractile function. The authors, therefore, reasoned that chronic dyssychrony of the cardiomyocyte circadian clock with its environment would precipitate myocardial maladaptation to a circadian challenge (simulated shiftwork; SSW). To test this hypothesis, 2- and 20-month-old wild-type and CCM (Cardiomyocyte Clock Mutant; a model with genetic temporal suspension of the cardiomyocyte circadian clock at the active-to-sleep phase transition) mice were subjected to chronic (16-wks) biweekly 12-h phase shifts in the LD cycle (i.e., SSW). Assessment of adaptation/maladaptation at whole-body homeostatic, gravimetric, humoral, histological, transcriptional, and cardiac contractile function levels revealed essentially identical responses between wild-type and CCM littermates. However, CCM hearts exhibited increased biventricular weight, cardiomyocyte size, and molecular markers of hypertrophy (anf, mcip1), independent of aging and/or SSW. Similarly, a second genetic model of selective temporal suspension of the cardiomyocyte circadian clock (Cardiomyocyte-specific BMAL1 Knockout [CBK] mice) exhibits increased biventricular weight and mcip1 expression. Wild-type mice exhibit 5-fold greater cardiac hypertrophic growth (and 6-fold greater anf mRNA induction) when challenged with the hypertrophic agonist isoproterenol at the active-to-sleep phase transition, relative to isoproterenol administration at the sleep-to-active phase transition. This diurnal variation was absent in CCM mice. Collectively, these data suggest that the cardiomyocyte circadian clock likely influences responsiveness of the heart to hypertrophic stimuli.
Collapse
Affiliation(s)
- David J Durgan
- Division of Cardiovascular Diseases, Department of Medicine, University of Alabama at Birmingham, 35294, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Nakazato R, Takarada T, Yamamoto T, Hotta S, Hinoi E, Yoneda Y. Selective upregulation of Per1 mRNA expression by ATP through activation of P2X7 purinergic receptors expressed in microglial cells. J Pharmacol Sci 2011; 116:350-61. [PMID: 21747211 DOI: 10.1254/jphs.11069fp] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Clock genes are believed to play a pivotal role in the generation and oscillation of circadian rhythm as a central clock in the hypothalamic suprachiasmatic nucleus in the mammalian brain. In this study, mRNA expression was for the first time demonstrated with clock genes in both cultured murine microglia and microglial cell line BV-2 cells. Exposure to ATP transiently increased Period-1 (Per1) mRNA expression without affecting that of other clock genes in BV-2 cells, while a similarly transient increase was shown in Per1 mRNA expression in a manner sensitive to P2X7 purinergic receptor antagonists in cultured microglia exposed to ATP. In BV-2 cells transfected with a Per1 promoter luciferase reporter plasmid, ATP significantly increased luciferase activity in a manner sensitive to a P2X7-receptor antagonist. In both microglia and BV-2 cells, a significant increase by ATP was seen in the immunocytochemical fluorescence intensity of cells expressing Per1 protein, with mRNA expression of different P2 receptors including P2X7. Per1 siRNA significantly decreased the number of cells with processes in BV-2 cells exposed to ATP. These results suggest that ATP selectively promotes Per1 expression through gene transactivation after stimulation of P2X7 purinergic receptors in microglial cells.
Collapse
Affiliation(s)
- Ryota Nakazato
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Natural Science and Technology, Japan
| | | | | | | | | | | |
Collapse
|
89
|
Portaluppi F, Tiseo R, Smolensky MH, Hermida RC, Ayala DE, Fabbian F. Circadian rhythms and cardiovascular health. Sleep Med Rev 2011; 16:151-66. [PMID: 21641838 DOI: 10.1016/j.smrv.2011.04.003] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 04/27/2011] [Indexed: 11/30/2022]
Abstract
The functional organization of the cardiovascular system shows clear circadian rhythmicity. These and other circadian rhythms at all levels of organization are orchestrated by a central biological clock, the suprachiasmatic nuclei of the hypothalamus. Preservation of the normal circadian time structure from the level of the cardiomyocyte to the organ system appears to be essential for cardiovascular health and cardiovascular disease prevention. Myocardial ischemia, acute myocardial infarct, and sudden cardiac death are much greater in incidence than expected in the morning. Moreover, supraventricular and ventricular cardiac arrhythmias of various types show specific day-night patterns, with atrial arrhythmias--premature beats, tachycardias, atrial fibrillation, and flutter - generally being of higher frequency during the day than night--and ventricular fibrillation and ventricular premature beats more common, respectively, in the morning and during the daytime activity than sleep span. Furthermore, different circadian patterns of blood pressure are found in arterial hypertension, in relation to different cardiovascular morbidity and mortality risk. Such temporal patterns result from circadian periodicity in pathophysiological mechanisms that give rise to predictable-in-time differences in susceptibility-resistance to cyclic environmental stressors that trigger these clinical events. Circadian rhythms also may affect the pharmacokinetics and pharmacodynamics of cardiovascular and other medications. Knowledge of 24-h patterns in the risk of cardiac arrhythmias and cardiovascular disease morbidity and mortality plus circadian rhythm-dependencies of underlying pathophysiologic mechanisms suggests the requirement for preventive and therapeutic interventions is not the same throughout the day and night, and should be tailored accordingly to improve outcomes.
Collapse
|
90
|
Takeda N, Maemura K. Circadian clock and cardiovascular disease. J Cardiol 2011; 57:249-56. [DOI: 10.1016/j.jjcc.2011.02.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Accepted: 02/22/2011] [Indexed: 12/17/2022]
|
91
|
Peliciari-Garcia RA, Zanquetta MM, Andrade-Silva J, Gomes DA, Barreto-Chaves ML, Cipolla-Neto J. Expression of circadian clock and melatonin receptors within cultured rat cardiomyocytes. Chronobiol Int 2011; 28:21-30. [PMID: 21182401 DOI: 10.3109/07420528.2010.525675] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Melatonin, the pineal gland hormone, provides entrainment of many circadian rhythms to the ambient light/dark cycle. Recently, cardiovascular studies have demonstrated melatonin interactions with many physiological processes and diseases, such as hypertension and cardiopathologies. Although membrane melatonin receptors (MT1, MT2) and the transcriptional factor RORα have been reported to be expressed in the heart, there is no evidence of the cell-type expressing receptors as well as the possible role of melatonin on the expression of the circadian clock of cardiomyocytes, which play an important role in cardiac metabolism and function. Therefore, the aim of this study was to evaluate the mRNA and protein expressions of MT1, MT2, and RORα and to determine whether melatonin directly influences expression of circadian clocks within cultured rat cardiomyocytes. Adult rat cardiomyocyte cultures were created, and the cells were stimulated with 1 nM melatonin or vehicle. Gene expressions were assayed by real-time polymerase chain reaction (PCR). The mRNA and protein expressions of membrane melatonin receptors and RORα were established within adult rat cardiomyocytes. Two hours of melatonin stimulation did not alter the expression pattern of the analyzed genes. However, given at the proper time, melatonin kept Rev-erbα expression chronically high, specifically 12 h after melatonin treatment, avoiding the rhythmic decline of Rev-erbα mRNA. The blockage of MT1 and MT2 by luzindole did not alter the observed melatonin-induced expression of Rev-erbα mRNA, suggesting the nonparticipation of MT1 and MT2 on the melatonin effect within cardiomyocytes. It is possible to speculate that melatonin, in adult rat cardiomyocytes, may play an important role in the light signal transduction to peripheral organs, such as the heart, modulating its intrinsic rhythmicity.
Collapse
Affiliation(s)
- Rodrigo A Peliciari-Garcia
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | | | | | | | | |
Collapse
|
92
|
Anigbogu CN, Williams DT, Brown DR, Silcox DL, Speakman RO, Brown LC, Karounos DG, Randall DC. Circadian Variations in Blood Pressure, Heart Rate, and HR-BP Cross-Correlation Coefficient during Progression of Diabetes Mellitus in Rat. Int J Hypertens 2011; 2011:738689. [PMID: 21629872 PMCID: PMC3095977 DOI: 10.4061/2011/738689] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Accepted: 02/10/2011] [Indexed: 11/20/2022] Open
Abstract
Circadian changes in cardiovascular function during the progression of diabetes mellitus in the diabetes prone rat (BBDP) (n = 8) were studied. Age-matched diabetes-resistant rats (BBDR) served as controls. BP was recorded via telemetry in contiguous 4 hr time periods over 24 hours starting with 12 midnight to 4 am as period zero (P0). Prior to onset of diabetes BP was high at P0, peaked at P2, and then fell again at P3; BP and heart rate (HR) then increased gradually at P4 and leveled off at P5, thereby exhibiting a bipodal rhythm. These patterns changed during long-term diabetes. The cross-correlation coefficient of BP and HR was not significantly different across groups at onset, but it fell significantly at 9 months of duration of diabetes (BBDP: 0.39 ± 0.06; BBDR: 0.65 ± 0.03; P < .05). These results show that changes in circadian cardiovascular rhythms in diabetes mellitus became significant at the late stage of the disease.
Collapse
Affiliation(s)
- Chikodi N. Anigbogu
- Department of Physiology, College of Medicine, University of Lagos, 12003 Lagos, Nigeria
- Department of Physiology, College of Medicine University of Kentucky, Lexington, KY 40536-0298, USA
| | - Daniel T. Williams
- Department of Physiology, College of Medicine University of Kentucky, Lexington, KY 40536-0298, USA
| | - David R. Brown
- Department of Physiology, College of Medicine University of Kentucky, Lexington, KY 40536-0298, USA
| | - Dennis L. Silcox
- Department of Physiology, College of Medicine University of Kentucky, Lexington, KY 40536-0298, USA
| | - Richard O. Speakman
- Department of Physiology, College of Medicine University of Kentucky, Lexington, KY 40536-0298, USA
| | - Laura C. Brown
- Department of Physiology, College of Medicine University of Kentucky, Lexington, KY 40536-0298, USA
| | - Dennis G. Karounos
- Lexington VA Medical Center and Department of Internal Medicine, College of Medicine University of Kentucky, Lexington, KY 40536-0298, USA
| | - David C. Randall
- Department of Physiology, College of Medicine University of Kentucky, Lexington, KY 40536-0298, USA
| |
Collapse
|
93
|
Ko ML, Shi L, Grushin K, Nigussie F, Ko GYP. Circadian profiles in the embryonic chick heart: L-type voltage-gated calcium channels and signaling pathways. Chronobiol Int 2011; 27:1673-96. [PMID: 20969517 DOI: 10.3109/07420528.2010.514631] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Circadian clocks exist in the heart tissue and modulate multiple physiological events, from cardiac metabolism to contractile function and expression of circadian oscillator and metabolic-related genes. Ample evidence has demonstrated that there are endogenous circadian oscillators in adult mammalian cardiomyocytes. However, mammalian embryos cannot be entrained independently to light-dark (LD) cycles in vivo without any maternal influence, but circadian genes are well expressed and able to oscillate in embryonic stages. The authors took advantage of using chick embryos that are independent of maternal influences to investigate whether embryonic hearts could be entrained under LD cycles in ovo. The authors found circadian regulation of L-type voltage-gated calcium channels (L-VGCCs), the ion channels responsible for the production of cardiac muscle contraction in embryonic chick hearts. The mRNA levels and protein expression of VGCCα1C and VGCCα1D are under circadian control, and the average L-VGCC current density is significantly larger when cardiomyocytes are recorded during the night than day. The phosphorylation states of several kinases involved in insulin signaling and cardiac metabolism, including extracellular signal-regulated kinase (Erk), stress-activated protein kinase (p38), protein kinase B (Akt), and glycogen synthase kinase-3β (GSK-3β), are also under circadian control. Both Erk and p38 have been implicated in regulating cardiac contractility and in the development of various pathological states, such as cardiac hypertrophy and heart failure. Even though both Erk and phosphoinositide 3-kinase (PI3K)-Akt signaling pathways participate in complex cellular processes regarding physiological or pathological states of cardiomyocytes, the circadian oscillators in the heart regulate these pathways independently, and both pathways contribute to the circadian regulation of L-VGCCs.
Collapse
Affiliation(s)
- Michael L Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | | | | | | | | |
Collapse
|
94
|
Sack MN. Caloric excess or restriction mediated modulation of metabolic enzyme acetylation-proposed effects on cardiac growth and function. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:1279-85. [PMID: 21295620 DOI: 10.1016/j.bbamcr.2011.01.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 01/19/2011] [Accepted: 01/26/2011] [Indexed: 12/16/2022]
Abstract
Caloric excess has been postulated to disrupt cardiac function via (i) the generation of toxic intermediates, (ii) via protein glycosylation and (iii) through the generation of reactive oxygen species. It is now increasingly being recognized that the nutrient intermediates themselves may modulate metabolic pathways through the post-translational modifications of metabolic enzymes. In light of the high energy demand of the heart, these nutrient mediated modulations in metabolic pathway functioning may play an important role in cardiac function and in the capacity of the heart to adapt to biomechanical stressors. In this review the role of protein acetylation and deacetylation in the control of metabolic programs is explored. Although not extensively investigated directly in the heart, the emerging data support that these nutrient mediated post-translational regulatory events (i) modulate cardiac metabolic pathways, (ii) integrate nutrient flux mediated post-translational effects with cardiac function and (iii) may be important in the development of cardiac pathology. Areas of investigation that need to be explored are highlighted. This article is part of a Special Issue entitled: Mitochondria and Cardioprotection.
Collapse
Affiliation(s)
- Michael N Sack
- Translational Medicine Branch, NHLBI, NIH, Bld 10-CRC, Room 5–3150, 10 Center Drive, Bethesda, MD, 20892-1454, USA.
| |
Collapse
|
95
|
Sachan N, Dey A, Rotter D, Grinsfelder DB, Battiprolu PK, Sikder D, Copeland V, Oh M, Bush E, Shelton JM, Bibb JA, Hill JA, Rothermel BA. Sustained hemodynamic stress disrupts normal circadian rhythms in calcineurin-dependent signaling and protein phosphorylation in the heart. Circ Res 2011; 108:437-45. [PMID: 21233454 DOI: 10.1161/circresaha.110.235309] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Despite overwhelming evidence of the importance of circadian rhythms in cardiovascular health and disease, little is known regarding the circadian regulation of intracellular signaling pathways controlling cardiac function and remodeling. OBJECTIVE To assess circadian changes in processes dependent on the protein phosphatase calcineurin, relative to changes in phosphorylation of cardiac proteins, in normal, hypertrophic, and failing hearts. METHODS AND RESULTS We found evidence of large circadian oscillations in calcineurin-dependent activities in the left ventricle of healthy C57BL/6 mice. Calcineurin-dependent transcript levels and nuclear occupancy of the NFAT (nuclear factor of activated T cells) regularly fluctuated as much as 20-fold over the course of a day, peaking in the morning when mice enter a period of rest. Phosphorylation of the protein phosphatase 1 inhibitor 1 (I-1), a direct calcineurin substrate, and phospholamban, an indirect target, oscillated directly out of phase with calcineurin-dependent signaling. Using a surgical model of cardiac pressure overload, we found that although calcineurin-dependent activities were markedly elevated, the circadian pattern of activation was maintained, whereas, oscillations in phospholamban and I-1 phosphorylation were lost. Changes in the expression of fetal gene markers of heart failure did not mirror the rhythm in calcineurin/NFAT activation, suggesting that these may not be direct transcriptional target genes. Cardiac function in mice subjected to pressure overload was significantly lower in the morning than in the evening when assessed by echocardiography. CONCLUSIONS Normal, opposing circadian oscillations in calcineurin-dependent activities and phosphorylation of proteins that regulate contractility are disrupted in heart failure.
Collapse
Affiliation(s)
- Nita Sachan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Szántóová K, Zeman M, Veselá A, Herichová I. Effect of phase delay lighting rotation schedule on daily expression of per2, bmal1, rev-erbα, pparα, and pdk4 genes in the heart and liver of Wistar rats. Mol Cell Biochem 2010; 348:53-60. [DOI: 10.1007/s11010-010-0636-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 10/28/2010] [Indexed: 01/21/2023]
|
97
|
Dominguez-Rodriguez A, Abreu-Gonzalez P, Sanchez-Sanchez JJ, Kaski JC, Reiter RJ. Melatonin and circadian biology in human cardiovascular disease. J Pineal Res 2010; 49:14-22. [PMID: 20536686 DOI: 10.1111/j.1600-079x.2010.00773.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Diurnal rhythms influence cardiovascular physiology, i.e. heart rate and blood pressure, and they appear to also modulate the incidence of serious adverse cardiac events. Diurnal variations occur also at the molecular level including changes in gene expression in the heart and blood vessels. Moreover, the risk/benefit ratio of some therapeutic strategies and the concentration of circulating cardiovascular system biomarkers may also vary across the 24-hr light/dark cycle. Synchrony between external and internal diurnal rhythms and harmony among molecular rhythms within the cell are essential for normal organ biology. Diurnal variations in the responsiveness of the cardiovascular system to environmental stimuli are mediated by a complex interplay between extracellular (i.e. neurohumoral factors) and intracellular (i.e. specific genes that are differentially light/dark regulated) mechanisms. Neurohormones, which are particularly relevant to the cardiovascular system, such as melatonin, exhibit a diurnal variation and may play a role in the synchronization of molecular circadian clocks in the peripheral tissue and the suprachiasmatic nucleus. Moreover, mounting evidence reveals that the blood melatonin rhythm has a crucial role in several cardiovascular functions, including daily variations in blood pressure. Melatonin has antioxidant, anti-inflammatory, chronobiotic and, possibly, epigenetic regulatory functions. This article reviews current knowledge related to the biological role of melatonin and its circadian rhythm in cardiovascular disease.
Collapse
|
98
|
Takeda N, Maemura K. Cardiovascular disease, chronopharmacotherapy, and the molecular clock. Adv Drug Deliv Rev 2010; 62:956-66. [PMID: 20451570 DOI: 10.1016/j.addr.2010.04.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Revised: 03/10/2010] [Accepted: 04/28/2010] [Indexed: 10/19/2022]
Abstract
Cardiovascular functions such as heart rate and blood pressure show 24h variation. The incidence of cardiovascular diseases including acute myocardial infarction and arrhythmia also exhibits diurnal variation. The center of this circadian clock is located in the suprachiasmatic nucleus in the hypothalamus. However, recent findings revealed that each organ, including cardiovascular tissues, has its own internal clock, which has been termed a peripheral clock. The functional roles played by peripheral clocks have been reported recently. Since the peripheral clock is considered to play considerable roles in the processes of cardiac tissues, the identification of genes specifically regulated by this clock will provide insights into its role in the pathogenesis of cardiovascular disorders. In addition, the discovery of small compounds that modulate the peripheral clock will help to establish chronotherapeutic approaches. Understanding the biological relevance of the peripheral clock will provide novel approaches to the prevention and treatment of cardiovascular diseases.
Collapse
|
99
|
Suzuki JI, Ogawa M, Tamura N, Maejima Y, Takayama K, Maemura K, Honda K, Hirata Y, Nagai R, Isobe M. A critical role of sympathetic nerve regulation for the treatment of impaired daily rhythm in hypertensive Dahl rats. Hypertens Res 2010; 33:1060-5. [PMID: 20668456 DOI: 10.1038/hr.2010.125] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
There is a deep relationship between impaired circadian rhythm and hypertension. However, the detailed mechanisms between the daily sleep-wake rhythm and cardiovascular disorders have not yet been elucidated. To clarify the mechanism, we examined salt-sensitive Dahl rats that were fed normal chow (n=10), high-salt chow (n=10) and high-salt chow with bisoprolol (n=10). Simultaneous electroencephalogram, electromyogram and locomotor activity were examined to analyze the sleep-wake state. We also examined heart rate, blood pressure and echocardiographic findings to verify the presence of hypertension. Hypertension with impaired ventricular contraction was observed in the rats with high-salt-chow consumption whereas normal-chow rats did not show these disorders. Although rats with the normal diet showed a standard daily rhythm with normal rapid eye movement (REM) sleep duration and locomotor activity, the high-salt-diet group exhibited an impaired daily rhythm with suppressed REM sleep and significant abnormal locomotor activity. Bisoprolol significantly improved the daily sleep-wake rhythm and locomotor activity. We showed that an impaired daily rhythm was closely related to the development of hypertension. Regulation of sympathetic nerve alterations may have a key role in the treatment of hypertension and circadian rhythm disorder.
Collapse
Affiliation(s)
- Jun-ichi Suzuki
- Department of Advanced Clinical Science and Therapeutics, University of Tokyo, 7-3-1 Hongo, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Zhou L, Zhang P, Cheng Z, Hao W, Wang R, Fang Q, Cao JM. Altered circadian rhythm of cardiac β3-adrenoceptor activity following myocardial infarction in the rat. Basic Res Cardiol 2010; 106:37-50. [PMID: 20661603 DOI: 10.1007/s00395-010-0110-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 07/06/2010] [Accepted: 07/09/2010] [Indexed: 01/03/2023]
Abstract
Circadian rhythms influence the incidence of adverse cardiac events but the underlying mechanisms are not well defined. We sought to investigate the role of the β3-adrenoceptor (β3-AR) in cardiac circadian disorders and arrhythmia severity after myocardial infarction (MI). MI was created by ligating the left anterior descending coronary artery of the rat heart in situ. Circadian variations of the myocardial expressions of β3-AR and clock genes Bmal1 and Npas2 were examined by real time reverse transcription polymerase chain reaction, Western blot and immunohistochemistry. Electrocardiograms and myocardial contraction were recorded in vivo and/or ex vivo. Ventricular tachyarrhythmias were induced by isoprenaline. Normal rats showed circadian oscillations in both the myocardial transcriptional expression of β3-AR and the β3-AR-induced positive chronotropic and negative inotropic cardiac effects. However, these circadian rhythms were significantly blunted or even abolished in rats with either acute MI (within 24 h) or healed MI (14 days after coronary ligation). The nocturnal level of β3-AR protein was higher in MI rats than in normal rats. In contrast, the circadian oscillations of the transcripts of Bmal1 and Npas2 in the myocardium were significantly augmented in rats with either acute MI or healed MI. BRL37344, a preferential β3-AR selective agonist, reduced the occurrence of ventricular tachycardia (VT) and ventricular fibrillation (VF) in rats with either acute MI or healed MI. We conclude that circadian rhythms of myocardial β3-AR activities are disturbed after MI and β3-AR activation offers anti-arrhythmic protection.
Collapse
Affiliation(s)
- Lan Zhou
- Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China
| | | | | | | | | | | | | |
Collapse
|