Elsevier

Sleep Medicine Reviews

Volume 16, Issue 2, April 2012, Pages 151-166
Sleep Medicine Reviews

Clinical Review
Circadian rhythms and cardiovascular health

https://doi.org/10.1016/j.smrv.2011.04.003Get rights and content

Summary

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.

Introduction

Biological processes and functions oscillate rhythmically in time. Circadian rhythms, which are of particular relevance to everyday life and clinical medicine, are controlled by an inherited master clock residing in the paired suprachiasmatic nuclei (SCN) of the hypothalamus.1 Rhythmic activities the SCN clock genes Per1, Per2, Per3, Bmal, Clock, and Cry and their gene products comprise the central time-keeping mechanism. Transcription factors CLOCK and BMAL1 drive the expression of Per1, Per2, Cry1, Cry2, plus a variety of clock-controlled genes, i.e., target genes that are not integral clock components and that do not feedback on CLOCK/BMAL1, via E-box sequences in their promoters. PER and CRY proteins negatively feedback on the transcriptional activity of CLOCK:BMAL1, which results in a circadian rhythm in expression of the CLOCK:BMAL1-driven clock and various clock-controlled genes. Precision of the period and staging of circadian rhythms is achieved via cyclic environmental time cues. The ambient light–dark cycle is the most important cue under normal conditions, with the retinohypothalamic neural projection relaying information sensed by specialized non-cone and non-rod receptors of the retina to the SCN about the timing of light onsets and offsets during each 24-h period. The biological time-keeping system also includes the multitude of peripheral cell, tissue, and organ circadian clocks that are regulated and coordinated by the master SCN clock. The output of the central and peripheral circadian clocks is mediated by various clock-controlled genes, giving rise to the body’s circadian time structure (CTS) that is appropriately staged by environmental time cues to support optimal human metabolic and performance efficiency and capability during diurnal activity and repair and rejuvenation during nighttime rest/sleep.

Circadian clocks have been identified in nearly all mammalian cells investigated, including cardiomyocytes, fibroblasts, vascular smooth muscle cells, and endothelial cells.2, 3, 4, 5, 6 Although 24-h rhythms in heart rate (HR), blood pressure (BP), and cardiac output are classically attributed to rhythms in neuroendocrine constituents, rhythms at the cellular level also play an important role.7, 8, 9 The evidence for this comes from a series of different studies. Genetic manipulation of circadian clock components, such as CLOCK and BMAL1,10 variations within a tandem repeat of the human clock gene Per3,11 and genetic ablation of the circadian clock within endothelial or vascular smooth muscle cells,12 either alter, markedly attenuate, or abolish HR and/or BP circadian rhythms. Furthermore, studies on the cardiomyocyte-specific clock mutant mouse model, in which the cardiomyocyte circadian clock is temporally locked to the commencement of the inactive/sleep phase, show the cardiomyocyte clock differentially regulates cardiac metabolism and contractile function during the 24 h.13, 14, 15, 16 Furthermore, rodent models reveal it directly modulates myocardial ischemia/reperfusion tolerance in a circadian rhythm-dependent manner.14 Recent findings also implicate the disruption (desynchronization) of circadian clocks in the pathogenesis of cardiovascular disease (CVD).8, 17 Circadian clocks are altered in numerous animal models of increased CVD risk, including aging, diet-induced obesity, diabetes mellitus, hypertension- and pressure overload-induced hypertrophy, simulated shift work, and ischemia/reperfusion.18, 19, 20, 21, 22, 23, 24 Circadian desynchrony of the organism from its environment, either in humans through rotating shift-work schedules or in rodent through light/dark cycle or genetic alteration, augments CVD development.25, 26, 27, 28 Furthermore, rodent studies show chronic desynchrony of the circadian clock results in enhancement of cardiac mass, cardiomyocyte size, and expressed hypertrophic markers, thereby suggesting it influences responsiveness to pro-hypertrophic stimuli.29

Beyond the cellular level, circadian rhythms of cardiovascular physiology and function are very well established30; moreover, clear circadian rhythmicity is found in pathophysiological mechanisms that underlie 24-h patterning of morbid and mortal CVD events. Moreover, the administration time, relative to the staging of involved circadian rhythms, of various classes of medications used to manage CVD risk may impact, sometimes quite dramatically, their pharmacokinetics (PK) and pharmacodynamics (PD).31 This infers it is necessary to tailor preventive and therapeutic interventions to circadian rhythm determinants to optimize intended outcomes.32

Recognition of the importance of circadian rhythms in cardiovascular functions and their involvement in 24-h patterns of CVD conditions and events has lead to renewed scientific interest in chronobiology, i.e., the study of biological rhythms. Indeed, investigation of the temporal structure of the sources and mechanisms of cardiovascular rhythms is a necessary preliminary step in understanding their clinical implications, especially in regard to CVD risk and its control through therapeutic interventions. The main sources of the time-dependency of cardiovascular physiology and pathophysiology are cyclic variation in external stimuli and demands, especially physical and mental activity and stress, and endogenous circadian rhythms, even though it is often impossible to clearly separate the relative contribution of each. This article discusses the relevance of circadian rhythms to the prevention and management of CVD, focusing on the three major clinical entities of arterial hypertension, myocardial ischemia (MI), and cardiac arrhythmias. Published pathophysiologic, epidemiologic, and clinico-therapeutic evidence clearly establishes the importance of a chronobiologic approach, both to uncovering new insight into the maintenance of cardiovascular health and to improving the prognostic assessment and therapeutic management of CVD patients.

Section snippets

Circadian rhythms in arterial hypertension

Systolic (SBP) and diastolic (DBP) BP exhibit distinct, although different, 24-h patterning among patients. In so-called normal dippers, the sleep-time BP mean is reduced by 10–20% relative to the daytime mean.33 During the daytime, typically two daytime peaks are manifested, the first more prominent one shortly after morning awakening and the second late in the afternoon or early evening, with a small mid-afternoon nadir. In healthy young adults, the immediate morning SBP rise amounts to about

Circadian changes in the pathophysiological mechanisms of arterial hypertension

The 24-h BP variation results primarily from the cyclic exogenous alteration of physical and mental activity, stress, among others, coupled with the sleep–wake cycle. However, endogenous neurohumoral and other circadian rhythms also play a role,75 although it is impossible to clearly separate the relative influence of the former form the latter. For sure, the effects of physical and mental activity account for the predominant proportion of the day-night variation,76 as demonstrated by studies

Twenty-four-hour pattern in MI

MI is the underlying pathogenetic mechanism of multiple clinical manifestations of CVD events: transient ischemic episodes, acute myocardial infarction (AMI), and sudden cardiac death (SCD). MI may result from either restricted and insufficient oxygen supply or increased oxygen demand. A number of physiologic variables are crucial in determining mismatch between myocardial oxygen supply and demand, and predictable circadian changes are exhibited by all of them.32 Such predictable-in-time

Circadian changes in the pathophysiological mechanisms of MI

MI is thought to be triggered by several pathophysiological mechanisms, particularly the sudden morning increase of BP, HR, SNS activity, basal vascular tone, vasoconstrictive hormones, prothrombotic tendency, platelet aggregability, plasma viscosity, and hematocrit.198 Each of these variables exhibits prominent circadian rhythmicity in phase with the reported 24-h pattern of the ischemic events.

Circadian rhythms in arrhythmias

Circadian variations have been established also in the pattern of presentation of both supraventricular216, 217 and ventricular cardiac arrhythmias,218, 219, 220 irrespective of the presence or absence of concomitant medications.221 In clinical practice, many arrhythmic episodes are observed as a consequence of MI, a pathophysiologic event that exhibits profound 24-h patterning, as previously related. However, the distribution during the 24 h of malignant arrhythmias is similar in patients with

Circadian changes in the pathophysiological mechanisms of arrhythmias

Many functional, e.g., neurohumoral, electrolytic, hemodynamic, metabolic, etc., factors, aside from MI, may trigger and maintain arrhythmic episodes. Mechanisms by which these factors interact to determine an arrhythmogenic stimulus remain incompletely understood, constituting a major problem not only for the characterization of the determinants of the 24-h pattern of arrhythmias, but also for devising optimal conventional or chronotherapeutic and chronopreventive strategies. Arrhythmias are

Perspectives and conclusions

The purpose of this review has been to highlight the (chrono)epidemiology and chronobiology of CVD and potential underlying pathogenic mechanisms. Clinically significant 24-h patterns in hypertensive BP levels, MI, AMI, SCD, and various atrial and ventricular arrhythmias are known, having been established by clinical case series and population-based studies plus meta-analysis of the data of individual reports. Clinical epidemiologists initially emphasized environmental factors as triggers of

References* (300)

  • A. Knutsson et al.

    Increased risk of ischaemic heart disease in shift workers

    Lancet

    (1986)
  • F. Portaluppi et al.

    Time-dependent effect of isradipine on the nocturnal hypertension of chronic renal failure

    Am J Hypertens

    (1995)
  • M.L. Tuck et al.

    Enhanced 24-hour norepinephrine and renin secretion in young patients with essential hypertension: relation with the circadian pattern of arterial blood pressure

    Am J Cardiol

    (1985)
  • P. Broadhurst et al.

    Ambulatory intra-arterial blood pressure in normal subjects

    Am Heart J

    (1990)
  • M. Tominaga et al.

    Disparate circadian variations of blood pressure and body temperature in bedridden elderly patients with cerebral atrophy

    Am J Hypertens

    (1995)
  • S.S. Franklin et al.

    Relationship between arterial blood pressure and plasma norepinephrine levels in a patient with neurogenic hypertension

    Am J Med

    (1986)
  • D.B. Shaw et al.

    Variations in blood pressure in hypertensives during sleep

    Lancet

    (1963)
  • A. de la Sierra et al.

    Assessment of salt sensitivity in essential hypertension by 24-h ambulatory blood pressure monitoring

    Am J Hypertens

    (1995)
  • M.P. Caruana et al.

    Effects of chronic congestive heart failure secondary to coronary artery disease on the circadian rhythm of blood pressure and heart rate

    Am J Cardiol

    (1988)
  • D. Sander et al.

    Circadian blood pressure patterns in four cases with hemodynamic brain infarction and prolonged blood–brain barrier disturbance

    Clin Neurol Neurosurg

    (1993)
  • L.A. Clark et al.

    A quantitative analysis of the effects of activity and time of day on the diurnal variations of blood pressure

    J Chronic Dis

    (1987)
  • C. Fava et al.

    Dipping and variability of blood pressure and heart rate at night are heritable traits

    Am J Hypertens

    (2005)
  • B.J. Janssen et al.

    Suprachiasmatic lesions eliminate 24-h blood pressure variability in rats

    Physiol Behav

    (1994)
  • M.H. Smolensky et al.

    Role of sleep-wake cycle on blood pressure circadian rhythms and hypertension

    Sleep Med

    (2007)
  • G. Coccagna et al.

    Laboratory note. Arterial pressure changes during spontaneous sleep in man

    Electroencephalogr Clin Neurophysiol

    (1971)
  • E. Lugaresi et al.

    Some periodic phenomena arising during drowsiness and sleep in man

    Electroencephalogr Clin Neurophysiol

    (1972)
  • D.A. Kirby et al.

    Differential effects of sleep stage on coronary hemodynamic function during stenosis

    Physiol Behav

    (1989)
  • D. Duguay et al.

    The crosstalk between physiology and circadian clock proteins

    Chronobiol Int

    (2009)
  • D.J. Durgan et al.

    The intrinsic circadian clock within the cardiomyocyte

    Am J Physiol Heart Circ Physiol

    (2005)
  • H. Nonaka et al.

    Angiotensin II induces circadian gene expression of clock genes in cultured vascular smooth muscle cells

    Circulation

    (2001)
  • G.K. Paschos et al.

    Circadian clocks and vascular function

    Circ Res

    (2010)
  • R.D. Rudic et al.

    Pressed for time: the circadian clock and hypertension

    J Appl Physiol

    (2009)
  • M.E. Young

    The circadian clock within the heart: potential influence on myocardial gene expression, metabolism, and function

    Am J Physiol Heart Circ Physiol

    (2006)
  • A.M. Curtis et al.

    Circadian variation of blood pressure and the vascular response to asynchronous stress

    Proc Natl Acad Sci USA

    (2007)
  • A.U. Viola et al.

    PER3 polymorphism and cardiac autonomic control: effects of sleep debt and circadian phase

    Am J Physiol Heart Circ Physiol

    (2008)
  • M.S. Bray et al.

    Disruption of the circadian clock within the cardiomyocyte influences myocardial contractile function, metabolism, and gene expression

    Am J Physiol Heart Circ Physiol

    (2008)
  • D.J. Durgan et al.

    Short communication: ischemia/reperfusion tolerance is time-of-day-dependent: mediation by the cardiomyocyte circadian clock

    Circ Res

    (2010)
  • M.E. Young

    Anticipating anticipation: pursuing identification of cardiomyocyte circadian clock function

    J Appl Physiol

    (2009)
  • T. Kunieda et al.

    Cellular senescence impairs circadian expression of clock genes in vitro and in vivo

    Circ Res

    (2006)
  • T. Mohri et al.

    Alterations of circadian expressions of clock genes in Dahl salt-sensitive rats fed a high-salt diet

    Hypertension

    (2003)
  • M.E. Young et al.

    Clock genes in the heart: characterization and attenuation with hypertrophy

    Circ Res

    (2001)
  • T.A. Martino et al.

    Disturbed diurnal rhythm alters gene expression and exacerbates cardiovascular disease with rescue by resynchronization

    Hypertension

    (2007)
  • T.A. Martino et al.

    Circadian rhythm disorganization produces profound cardiovascular and renal disease in hamsters

    Am J Physiol Regul Integr Comp Physiol

    (2008)
  • M.J. Sole et al.

    Diurnal physiology: core principles with application to the pathogenesis, diagnosis, prevention, and treatment of myocardial hypertrophy and failure

    J Appl Physiol

    (2009)
  • D.J. Durgan et al.

    Evidence suggesting that the cardiomyocyte circadian clock modulates responsiveness of the heart to hypertrophic stimuli in mice

    Chronobiol Int

    (2011)
  • F. Portaluppi et al.

    Circadian rhythm and environmental determinants of blood pressure regulation in normal and hypertensive conditions

  • F. Portaluppi et al.

    From a static to a dynamic concept of risk: the circadian epidemiology of cardiovascular events

    Chronobiol Int

    (1999)
  • F. Snyder et al.

    Changes in respiration, heart rate and systolic blood pressure in human sleep

    J Appl Physiol

    (1964)
  • A. Reinberg et al.

    Rythmes circadiens du pouls, de la pression arterielle, des excretions urinaires en 17-hydroxycorticosteroides catecholamines et potassium chez l’homme adulte sain, actif et au repos

    Ann Endocrinol (Paris)

    (1970)
  • A.B. Davies et al.

    Circadian rhythm of blood pressure in patients dependent on ventricular demand pacemakers

    Br Heart J

    (1984)
  • Cited by (228)

    View all citing articles on Scopus
    *

    The most important references are denoted by an asterisk.

    View full text