13
Review Consequences of Circadian and Sleep Disturbances for the Cardiovascular System Faisal J. Alibhai, BSc, Elena V. Tsimakouridze, BSc, Cristine J. Reitz, BSc, W. Glen Pyle, PhD, and Tami A. Martino, PhD Cardiovascular Research Group, Department of Biomedical Sciences, University of Guelph, Ontario, Canada ABSTRACT Circadian rhythms play a crucial role in our cardiovascular system. Importantly, there has been a recent urry of clinical and experimental studies revealing the profound adverse consequences of disturbing these rhythms on the cardiovascular system. For example, circadian disturbance worsens outcome after myocardial infarction with impli- cations for patients in acute care settings. Moreover, disturbing rhythms exacerbates cardiac remodelling in heart disease models. Also, circadian dyssynchrony is a causal factor in the pathogenesis of heart disease. These discoveries have profound implications for the cardiovascular health of shift workers, individuals with circadian and sleep disorders, or anyone subjected to the 24/7 demands of society. Moreover, these studies give rise to 2 new frontiers for translational research: (1) circadian rhythms and the cardiac sarcomere, which sheds new light on our understanding of myolament structure, sig- nalling, and electrophysiology; and (2) knowledge translation, which includes biomarker discovery (chronobiomarkers), timing of therapies (chronotherapy), and other new promising approaches to improve the management and treatment of cardiovascular disease. Reconsidering circadian rhythms in the clinical setting benets repair mechanisms, and offers new promise for patients. R ESUM E Les rythmes circadiens jouent un rôle crucial dans notre système cardiovasculaire. Notamment, une r ecente avalanche d etudes cliniques et exp erimentales r ev elant les cons equences ind esirables profondes de la perturbation de ces rythmes sur le système car- diovasculaire ont et er ealis ees. Par exemple, la perturbation des rythmes circadiens d et eriore les r esultats cliniques après linfarctus du myocarde des patients en soins de phase aiguë. De plus, la pertur- bation des rythmes exacerbe le processus de remodelage cardiaque des modèles de cardiopathie. Aussi, la dyssynchronie circadienne est un facteur causal dans la pathogenèse de la cardiopathie. Ces d ecouvertes ont de profondes cons equences sur la sant e car- diovasculaire des travailleurs de quart, des individus ayant des trou- bles du rythme circadien et du sommeil, ou de tout individu soumis tous les jours 24 heures sur 24 aux exigences de la soci et e. En outre, ces etudes mettent en exergue 2 nouvelles frontières de la recherche translationnelle : 1) les rythmes circadiens et le sarcomère cardiaque, lequel jette un nouvel eclairage sur notre compr ehension de la struc- ture des myolaments, de la signalisation et de l electrophysiologie; 2) lapplication des connaissances, qui comprend la d ecouverte des bio- marqueurs (chronobiomarqueurs), le meilleur moment des th erapies (chronoth erapie) et dautres nouvelles approches prometteuses pour am eliorer la prise en charge et le traitement de la maladie car- diovasculaire. La reconsid eration des rythmes circadiens en milieu clinique favorise les m ecanismes de r eparation et savère prometteuse pour les patients. Jürgen Aschoff was a physician, scientist, and cofounder of the eld of circadian biology. He introduced the notion that day and night (diurnal) rhythms in physiology are a fundamental feature in most living organisms including humans. 1 We now know that rhythms are regulated by the circadian system, by orchestration in the hypothalamic suprachiasmatic nucleus, and through neural and hormonal outputs to all organs including the heart (Fig. 1A). 2-7 Mechanistically, an exquisite underlying cellular clock mechanism has been identied, which controls molecular rhythms in virtually all cells including car- diomyocytes (Fig. 1B). 8,9 Over the past 5 decades numerous clinical and experimental studies have revealed a crucial role for the circadian system in regulating healthy physiology. Most recently, studies by our group and others have shed light on the profound adverse health consequences of circadian distur- bances, underlying cardiovascular, cancer, metabolic, respira- tory, psychiatric, and other disorders. 6,10-15 In this review we focus on the consequences of circadian disturbances on the pathogenesis and pathophysiology of heart disease. Canadian Journal of Cardiology 31 (2015) 860e872 Received for publication October 31, 2014. Accepted January 8, 2015. Corresponding author: Dr Tami A. Martino, Cardiovascular Research Group, Biomedical Sciences/OVC Room 1646B, University of Guelph, Ontario N1G2W1, Canada. Tel.: þ1-519-824-4120 54910; fax: þ1-519- 767-1450. E-mail: [email protected] See page 867 for disclosure information. http://dx.doi.org/10.1016/j.cjca.2015.01.015 0828-282X/Ó 2015 Canadian Cardiovascular Society. Published by Elsevier Inc. All rights reserved.

Consequences of Circadian and Sleep Disturbances for the

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Consequences of Circadian and Sleep Disturbances for the

rdiology 31 (2015) 860e872

Canadian Journal of Ca

Review

Consequences of Circadian and Sleep Disturbances forthe Cardiovascular System

Faisal J. Alibhai, BSc, Elena V. Tsimakouridze, BSc, Cristine J. Reitz, BSc, W. Glen Pyle, PhD,

and Tami A. Martino, PhDCardiovascular Research Group, Department of Biomedical Sciences, University of Guelph, Ontario, Canada

ABSTRACTCircadian rhythms play a crucial role in our cardiovascular system.Importantly, there has been a recent flurry of clinical and experimentalstudies revealing the profound adverse consequences of disturbingthese rhythms on the cardiovascular system. For example, circadiandisturbance worsens outcome after myocardial infarction with impli-cations for patients in acute care settings. Moreover, disturbingrhythms exacerbates cardiac remodelling in heart disease models.Also, circadian dyssynchrony is a causal factor in the pathogenesis ofheart disease. These discoveries have profound implications for thecardiovascular health of shift workers, individuals with circadian andsleep disorders, or anyone subjected to the 24/7 demands of society.Moreover, these studies give rise to 2 new frontiers for translationalresearch: (1) circadian rhythms and the cardiac sarcomere, whichsheds new light on our understanding of myofilament structure, sig-nalling, and electrophysiology; and (2) knowledge translation, whichincludes biomarker discovery (chronobiomarkers), timing of therapies(chronotherapy), and other new promising approaches to improve themanagement and treatment of cardiovascular disease. Reconsideringcircadian rhythms in the clinical setting benefits repair mechanisms,and offers new promise for patients.

Received for publication October 31, 2014. Accepted January 8, 2015.

Corresponding author: Dr Tami A. Martino, Cardiovascular ResearchGroup, Biomedical Sciences/OVC Room 1646B, University of Guelph,Ontario N1G2W1, Canada. Tel.: þ1-519-824-4120 �54910; fax: þ1-519-767-1450.

E-mail: [email protected] page 867 for disclosure information.

http://dx.doi.org/10.1016/j.cjca.2015.01.0150828-282X/� 2015 Canadian Cardiovascular Society. Published by Elsevier Inc. A

R�ESUM�ELes rythmes circadiens jouent un rôle crucial dans notre systèmecardiovasculaire. Notamment, une r�ecente avalanche d’�etudescliniques et exp�erimentales r�ev�elant les cons�equences ind�esirablesprofondes de la perturbation de ces rythmes sur le système car-diovasculaire ont �et�e r�ealis�ees. Par exemple, la perturbation desrythmes circadiens d�et�eriore les r�esultats cliniques après l’infarctus dumyocarde des patients en soins de phase aiguë. De plus, la pertur-bation des rythmes exacerbe le processus de remodelage cardiaquedes modèles de cardiopathie. Aussi, la dyssynchronie circadienne estun facteur causal dans la pathogenèse de la cardiopathie. Cesd�ecouvertes ont de profondes cons�equences sur la sant�e car-diovasculaire des travailleurs de quart, des individus ayant des trou-bles du rythme circadien et du sommeil, ou de tout individu soumistous les jours 24 heures sur 24 aux exigences de la soci�et�e. En outre,ces �etudes mettent en exergue 2 nouvelles frontières de la recherchetranslationnelle : 1) les rythmes circadiens et le sarcomère cardiaque,lequel jette un nouvel �eclairage sur notre compr�ehension de la struc-ture des myofilaments, de la signalisation et de l’�electrophysiologie; 2)l’application des connaissances, qui comprend la d�ecouverte des bio-marqueurs (chronobiomarqueurs), le meilleur moment des th�erapies(chronoth�erapie) et d’autres nouvelles approches prometteuses pouram�eliorer la prise en charge et le traitement de la maladie car-diovasculaire. La reconsid�eration des rythmes circadiens en milieuclinique favorise les m�ecanismes de r�eparation et s’avère prometteusepour les patients.

Jürgen Aschoff was a physician, scientist, and cofounder of thefield of circadian biology. He introduced the notion that dayand night (diurnal) rhythms in physiology are a fundamentalfeature in most living organisms including humans.1 We nowknow that rhythms are regulated by the circadian system, byorchestration in the hypothalamic suprachiasmatic nucleus,

and through neural and hormonal outputs to all organsincluding the heart (Fig. 1A).2-7 Mechanistically, an exquisiteunderlying cellular clock mechanism has been identified, whichcontrols molecular rhythms in virtually all cells including car-diomyocytes (Fig. 1B).8,9 Over the past 5 decades numerousclinical and experimental studies have revealed a crucial role forthe circadian system in regulating healthy physiology. Mostrecently, studies by our group and others have shed light on theprofound adverse health consequences of circadian distur-bances, underlying cardiovascular, cancer, metabolic, respira-tory, psychiatric, and other disorders.6,10-15 In this review wefocus on the consequences of circadian disturbances on thepathogenesis and pathophysiology of heart disease.

ll rights reserved.

Page 2: Consequences of Circadian and Sleep Disturbances for the

Figure 1. Circadian rhythms. (A) Schematic representation of the circadian hierarchical oscillator model and the outputs that influence cardio-vascular processes. The suprachiasmatic nucleus in the hypothalamus synchronizes peripheral organ clocks, including the heart, to entrain to the24-hour light-dark environment. (B) The molecular clock mechanism keeps 24-hour time via transcription-translation feedback loops. The primarynegative feedback loop consists of response element (RRE) driven Brain and Muscle ARNT Like 1: Circadian Locomotor Cycles Kaput (BMAL1:-CLOCK), which leads to Period (PER) and Cryptochrome (CRY) production, PER:CRY phosphorylation by Casein Kinase 1 d/ε (CKd/ε), and PER:CRYself-repression. BMAL1:CLOCK also drives Nuclear Receptor Subfamily 1, Group D, Member 1 (REV-ERBa) expression, which represses BMAL1.This mechanism regulates expression of clock-controlled genes (ccg) which regulate rhythmic biological processes.

Alibhai et al. 861Circadian Rhythm Disturbance and Heart Disease

Circadian Biology and Normal CardiovascularPhysiology

Circadian rhythms in heart rate and blood pressure

Circadian rhythms are important for daily cyclic variationin mammalian physiology, for example our heart rate (HR)is highest during wake and lowest during sleep.16 Bloodpressure (BP) also displays a daily rhythm, indeed, Floras andcolleagues documented for the first time the remarkable BPsurge that occurs at the time of waking and morning activ-ity.17 BP is highest in the morning and decreases progres-sively (approximately 10%) to reach a nadir during sleep.18

These rhythms are consistent with the diurnal biases ofour autonomic nervous system, which oscillate over 24-hourperiods.19 Thus our cardiovascular physiology exhibitsstriking time of day-dependent oscillations. Remarkably,diurnal variations in HR and cardiac contractility observedin vivo still persist ex vivo, for example, when rodent heartsare perfused on a Langendorff apparatus.13,20 Thus it is notjust the neurohormonal axes that contribute to time of daycardiac physiology, but also the intrinsic circadian propertiesof the heart. Over the past decade this has given rise tonumerous experimental studies that show a direct role forthe circadian mechanism on our daily cardiovascularphysiology, as is demonstrated experimentally in rats21-24

and mice.13,25,26

Daily timing of onset of adverse cardiovascular events

Adverse cardiovascular events are a leading cause of deathworldwide and do not occur at random times throughout theday. An early morning peak of acute myocardial infarction(MI) was first reported in 1985,27 and in the decades since,despite changes in lifestyle and advances in medicine, thepattern of morning excess of MI persists.28-33 A diurnalpattern also occurs for infarct size,34-36 stroke,37 angina,38

ventricular tachyarrhythmia,39-41 defibrillation energy re-quirements,42 ventricular refractoriness,43 and sudden cardiacdeath.44-46 Physiologically, sympathovagal balance is thought

to play a causal role, because patients with autonomic nervoussystem dysfunction do not exhibit the early morning peak intiming of onset of MI.47-49 Several studies have highlightedadditional factors, many under circadian control, which in-crease the risk of adverse cardiovascular events, includingdiurnal rhythms in platelet activity,50-53 thrombosis orthrombolysis,54-56 and endothelial function.57 Mechanisti-cally, a direct role for the circadian mechanism underlyingtime of day dependence has been demonstrated inhumans52,58 and animal models.34,59-61

Diurnal variations in cardiomyocyte metabolism

As described, cardiac function displays a time of dayeffect, driven in part by the sympathetic environment andthe neurohormonal milieu. However, at the myocyte level,there is considerable new evidence that intrinsic cellularproperties also occur in a circadian manner, as has beenexquisitely detailed in studies by Young and colleagues. Tosummarize, this is especially evident from the ex vivoperfused rat or mouse heart models, which demonstratepersistence of time of day variations in cardiac metabolism,even in the absence of neural and hormonal cues.20,62,63

Day/night variations in cardiac metabolism allow theheart to switch between different substrates (mainly glucoseand fatty acids) for energy sources depending on availabilityover the course of 24-hour periods.64,65 These intrinsiccardiomyocyte processes are facilitated in part by thecardiomyocyte-specific circadian clock mechanism.13,66

This circadian mechanism regulates messenger RNA(mRNA) and protein expression of key metabolic factors incardiomyocytes as described herein, thus controlling cellularprocesses in a time of day-dependent manner.

Circadian genomics

The heart is genetically a different organ in the day vs thenight. The first large-scale microarray study to demonstratethis revealed that approximately 8% of genes are rhythmic inmurine hearts under circadian (constant dark) conditions.67

Page 3: Consequences of Circadian and Sleep Disturbances for the

862 Canadian Journal of CardiologyVolume 31 2015

However, because humans (and medicine) exist in a diurnaland not circadian environment, we showed that approxi-mately 13% of genes in murine hearts are rhythmic undernormal day and night conditions.68 These genomic experi-ments recapitulate earlier findings of individual clock genecycling in polymerase chain reaction analyses,69 and more-over, identify many new rhythmic genes important for cardiacgrowth, renewal, transcription, translation, signalling path-ways, and metabolism.67,68 Our circadian heart microarraydata (and that of other researchers) are available through anopen access Web site created by the Hogenesch laboratory(Circadian Expression Profiles Data Base, Circa DB, http://bioinf.itmat.upenn.edu/circa/).70-73 Rhythmic cycling of thecore circadian mechanism genes has also been recentlydemonstrated in human heart tissue, in polymerase chainreaction analyses.74 Consistent with these observations is thepersistent 24-hour rhythmic oscillations in bioluminescence inheart and vascular tissue explants from transgenic rats inwhich luciferase expression is driven by the circadian mech-anism factor, Period 1 (Per1).75 Moreover, rhythmicity hasalso been shown to occur at the cellular level, in car-diomyocytes, vascular smooth muscle cells, endothelial cells,and fibroblasts in culture.76-80 It is worth noting that re-searchers generally perform rodent studies during the lightperiod, during rodent sleep time, and results are frequentlycompared with human wake time. These molecular studiesclearly demonstrate that timing of functional assessments ishighly critical in interpreting experimental findings. In sum-mary, the cell types in the heart possess circadian clockmechanisms that regulate a variety of clock-controlled outputgenes, which in turn influence the time of day processesessential for heart structure and function.

Circadian proteomics

Because it is the proteins and not directly the mRNA thatunderlies fundamental biochemical processes in cells, studieshave recently begun to explore circadian variation in thecellular proteome. We found that approximately 8% of thesoluble cardiac proteome exhibits diurnal variation in murineheart, using a large-scale 2-dimensional difference in gelelectrophoresis and mass spectrometry approach.81 Thecircadian mechanism plays a role in protein abundance in theheart; cardiomyocyte-specific Clock mutant mice have altereddiurnal abundance of proteins, including many rate-limitingenzymes crucial for vital metabolic pathways in the heart.81

In terms of how time of day protein abundance iscontrolled, we reported that approximately half of the diurnalvariation in the cardiac proteome can be accounted for byunderlying rhythmic changes at the mRNA level.81 Ourfindings are consistent with a previous report by Reddy et al.,who similarly observed approximately 50% of mRNA fluc-tuations underlying the hepatic proteome.82 Most recently,Luck et al. developed a statistical model that showed thatrhythmic production and degradation (half-life) at the mRNAand protein levels underlies daily protein abundance.83

Additional mechanisms that regulate diurnal molecularrhythms are glucocorticoid control of transcription,84,85 andtime of day variations in chromatin architecture.86 Thus, thereis a dynamic nature to the cardiac proteome, which underliesdiurnal variations in heart function.

Experimental Models of Circadian Disruptionand Consequences for the CardiovascularSystem

As highlighted in the preceding section, circadian rhythmsare fundamentally important for cardiovascular health. Theheart displays clear differences in time of day organ, cellular,and molecular physiology. Questions arise, therefore, as towhat happens when these rhythms are disturbed. Circadianrhythm disturbances can have multiple deleterious effects onorgan physiology, with profound clinical implications forcardiovascular and other diseases.6,10,87-89 In this section wefocus on experimental studies investigating the profoundadverse effects of circadian rhythm disturbance on the car-diovascular system.

Diurnal disturbance worsens outcome after MI

MI triggers a temporally orchestrated inflammatoryresponse designed to clear the infarct of dead cells and debris,and at the same time activates reparative pathways for fibrosisand scar formation.90-92 We postulated that the circadianmechanism plays a key role in immune cell recruitment andinfarct healing after MI. That there is daily cycling of pe-ripheral blood cells,93-96 diurnal reactivity of immuno-cytes,95,97 and a circadian mechanism in immune cells98-102

provides indirect support for this hypothesis. Our recentstudy directly recognized, to our knowledge for the first time,that the circadian mechanism plays a causal role in immu-nocyte recruitment to infarcted myocardium, and that dis-turbing this mechanism worsens outcome after MI.103 Micewere infarcted using left anterior descending coronary arteryligation (MI model), and randomized to either a normaldiurnal or disrupted light:dark environment for 5 days, andthen maintained in normal diurnal conditions. Short-termdiurnal disruption altered innate immune responses crucialfor scar formation, leading to maladaptive cardiac remodelling(Fig. 2). One of the most intriguing findings is a role for thecircadian mechanism factor CLOCK in recruiting innateimmune cells after MI. This study has clinical relevance,because circadian and sleep disruptions can impair recovery inmodern intensive care units.104-107 Reconsidering how wepractice medicine, by maintaining the diurnal environmentand patient biological rhythms, provides a promising non-pharmaceutical approach to improve patient outcomes inacute clinical care settings.108

Diurnal disturbance worsens cardiac remodelling

The adverse consequences of diurnal disturbance on pre-existing heart disease was first demonstrated by Turek andcolleagues, who revealed the deleterious effects on survival incardiomyopathic hamsters subjected to chronic shifts in thelight and dark cycle.109 We showed that diurnal rhythmdisturbance exacerbates cardiac remodelling in mice withpressure overload-induced cardiac hypertrophy; and impor-tantly, that the adverse remodelling only ceased when theanimals were returned to their normal diurnal cycle.110 Thus,altering the photoperiod to one in which animals cannotentrain alters the relationship between the light and dark cy-cle, endogenous physiology, and cardiac gene and proteinlevels, such that individuals are “out of sync” with theirenvironment. This has direct consequences on cardiac

Page 4: Consequences of Circadian and Sleep Disturbances for the

Figure 2. Short-term diurnal rhythm disturbance in the first few days after myocardial infarction (MI) significantly worsens remodelling and outcome.Mice (C57Bl/6) were subjected to left anterior descending coronary artery ligation (MI model) then randomized to either a normal 12-hour light-dark(L:D) environment (normal diurnal environment) or a disturbed L:D environment for 5 days (short-term diurnal rhythm and sleep disturbance). Afteronly 5 days, all animals were placed in normal 12-hour light-dark L:D environment for the duration of the experiment (8 weeks; 8w post-MI).Histologic analyses revealed that the disrupted-MI mice had greater left ventricular dilation and infarct expansion by 8 weeks after MI comparedwith the normal-MI mice (Masson’s Trichrome). Overall, we showed that short-term disturbance of circadian rhythms after MI alters immunocyterecruitment to infarcted myocardium and impairs healing; conversely, maintaining rhythms is crucial for benefiting wound healing and outcome.103

These preclinical findings offer promise for clinical translation, because maintaining diurnal rhythms in intensive and acute care units in the first fewdays after injury can benefit healing and outcome.

Alibhai et al. 863Circadian Rhythm Disturbance and Heart Disease

physiology and pathophysiology, exacerbating underlyingheart disease.

Circadian dyssynchrony causes heart disease

The important consequences of circadian dyssynchrony onthe heart are perhaps best exemplified by the þ/tau hamsterstudies. These animals carry a mutation in the circadian clockmechanism gene, casein kinase 1 epsilon, such that theyexhibit an internal circadian period of approximately 22hours, even though they live in a 24-hour world.111 Theþ/tau heterozygotes exhibit abnormal entrainment to the 24-hour diurnal environment (earlier onset and a more frag-mented activity profile),112 and reduced longevity, comparedwith their wild type littermates.113 In terms of cardiovascularconsequences, the Sole and Ralph laboratory demonstratedthat circadian disorganization in the þ/tau hamsters is a causalfactor in the pathogenesis of dilated cardiomyopathy.114

Conversely, if the animals are housed in diurnal cyclesappropriate for their genotype (22 hours), then rhythmsnormalize and the heart disease is prevented.114 Notably, thetau/tau homozygotes have a 20-hour period and do not

entrain to a 24-hour light and dark cycle and thus they do notdevelop heart disease.114 Thus, these studies demonstrate theimportance of synchrony between the intrinsic circadianperiod and the external light-dark environment, highlightingthat abnormal entrainment by circadian dyssynchrony playsan aetiologic role in the pathogenesis of heart disease.

Circadian gene mutations and heart disease

Additional lines of evidence further support the notion thatcircadian mechanism gene factors underlie cardiovascularpathophysiology; several recent studies are summarized herein.(1) Whole body brain and muscle ARNT-like 1 (BMAL1)-/-

knockout mice exhibit a loss of diurnal variation in HR andBP,26 and develop dilated cardiomyopathy.115 (2)Cardiomyocyte-specific Bmal1 knockout mice also developdilated cardiomyopathy even though the disturbance is only inthe heart.66,116 (3) In gene mapping studies it was found thatBmal1 associates with hypertension loci in spontaneouslyhypertensive rats.117 (4) In a similar light, BMAL1 (and alsoneuronal PAS domain protein 2 [Npas2]) polymorphisms areassociated with hypertension in humans.117,118 (5) Mice with

Page 5: Consequences of Circadian and Sleep Disturbances for the

864 Canadian Journal of CardiologyVolume 31 2015

simultaneous loss of the circadian output genes develop car-diac hypertrophy and left ventricular dysfunction.119 (6)Cardiomyocyte-specific circadian mutant mice exhibit blunt-ed diurnal HR variation, reduced responsiveness of the heartto workload changes, and altered cardiac metabolism.13,65,120

(7) Period 2 (Per2) mice have attenuated injury in the coro-nary artery ligation model.121 (8) Adenosine receptor A2b-mediated stabilization of Per2 is protective against ischemicheart disease in mice.122 Collectively, these studies are crucialfor demonstrating relevant relationships between the circadianmechanism and cardiovascular disease genesis and pathology.

Translational Consequences of Circadian andSleep Disruption on the Cardiovascular Systemin Humans

Many of the mechanistic studies described herein thatinvestigated the effects of circadian and sleep disturbances onthe cardiovascular system, were performed in rodent modelsand cell culture. However, these have important translationalapplications for human health and for clinical cardiology, asdescribed herein.

Shift work and heart disease

Circadian rhythm disturbances are relevant to humans. Forexample, the World Health Organization describes shift workas an occupational disturbance of circadian rhythms,123 andclinical studies support an association between shift work andheart disease. Mechanistically, shift work alters sympatheticand vagal autonomic activity,124 and diurnal BP profiles,125

which might contribute to cardiovascular disease. Furthersupporting this notion, Scheer et al. demonstrated experi-mentally that the adverse effects of circadian misalignment inhealthy humans can affect endocrine, metabolic, and auto-nomic predictors of cardiovascular (and other) risk.126

Epidemiologic studies further support a link betweenshift work and heart disease. For example, an increased riskof ischemic heart disease was demonstrated in a longitudinalstudy of 504 paper mill shift workers in Sweden.127

Moreover, an increased risk of coronary heart disease wasreported for US female shiftwork nurses.128 Also, anincreased risk for MI was shown in a meta-analysis of 34studies pertaining to shift workers and cardiovascular dis-ease.129 Furthermore, a greater incidence of atherosclerosiswas demonstrated in a cross-sectional examination of 2510shift workers in Germany.130 Last, severe hypertension wasdemonstrated in a longitudinal study of 6495 Japanese maleshift workers.131 Because millions of North American andother individuals engage in shift work or irregular workschedules at some point in their careers,132 it is importantto determine how atypical work schedules affect perfor-mance efficiency,133 and minimize the potential for adversehealth consequences.

Circadian rhythms and insufficient sleep

Circadian rhythm disturbances are also relevant throughtheir intersection with sleep because sleep is under bothcircadian and homeostatic control. Indeed, a role for circa-dian mechanism genetic factors on sleep has been recentlydiscovered in humans. For example, familial advanced phase

sleep syndrome is associated with a missense mutation in theclock component PER2, which alters the circadian period.134

These individuals go to sleep very early in the night time,and routinely wake up in the extreme early morning hours.Although a direct correlation between circadian factors thatinfluence sleep and heart disease in humans has not yet beendemonstrated, factors affecting sleep duration clearly have adirect effect on cardiovascular health. For example, in today’ssociety, 40% of North Americans do not achieve the rec-ommended 7 hours of sleep,135 and short (<5 hours) or long(>9 hours) sleep durations are associated with an increasedrisk of heart disease.136 These findings are consistent withearlier reports of increased all-cause mortality risk (includingischemic heart disease) in individuals with sleeping patternsother than 7-8 hours per night.137,138 In addition to circa-dian mutations that cause sleep changes, circadian-coupledfactors disturbed by inadequate sleep might mechanisticallycontribute to heart disease pathophysiology.139-141 Intrigu-ingly, there are molecular consequences on diurnal geneexpression in the heart with as little as one night of sleepdeprivation. This was demonstrated in mice after 3, 6, 9, and12 hours of sleep deprivation, which altered expression of2470 genes in the heart in microarray analyses.142

Chronotypes, social clocks, and light at night

Circadian rhythms in some physiologic functions varybetween humans (eg, core body temperature, melatoninphase), leading to classifications of early/morning or late/evening chronotypes.143 Intriguingly, chronotype might in-fluence cardiovascular disease, because evening types appearto be more at risk for cardiovascular changes (hypertension,HR, BP) compared with morning types.144 A later chro-notype is also associated with social jet lagdthe misalign-ment of biological and social clocks145dand prevalence ofsocial jet lag in apparently healthy individuals mightcontribute to cardiovascular risk.146 Light at night includingthat from light-emitting diodes in television and computerscreens has broad consequences for our health, and is thesubject several interesting recent reports.147,148 These studiesare relevant to contemporary society and shed new light onhow our circadian phenotypes, lifestyle choices, and thediurnal environment can affect cardiovascular health anddisease.

New Frontiers: Circadian Rhythms and theCardiac Sarcomere

In addition to the mentioned experimental and clinicaldiscoveries, there are new frontiers for circadian rhythmsresearch important to our understanding of the cardiovascularsystem. The first involves circadian rhythms and the cardiacsarcomere, which is the main contractile apparatus in theheart. These studies give rise to new understanding ofmyofilament structure, signalling, and electrophysiology, asdetailed herein.

Circadian rhythms and myofilament structure

Cardiac myofilaments mediate heart performance(Fig. 3),149-152 and recent studies indicate a role for thecircadian mechanism in affecting myofilament and thus

Page 6: Consequences of Circadian and Sleep Disturbances for the

Figure 3. The diurnal cardiac sarcomere. (A) The cardiac sarcomere, delineated by 2 Z-discs, sits at the functional centre of myocytes. (B) Car-diomyocyte contraction is a calcium-dependent interaction, with steric modulation by the troponin-tropomyosin complex, leading to myosin and actincross-bridges. ATP cleavage by myosin heads provides energy for the power stroke that pulls actin toward the centre of the cell and produces cellshortening. Cross-bridge cycling exhibits a circadian rhythm and disruption of CLOCK abolishes the diurnal cycling of cardiac myofilament activa-tion.81 (C) Key elements of the cardiac sarcomere.

Alibhai et al. 865Circadian Rhythm Disturbance and Heart Disease

cardiac function. For example, ex vivo rat hearts exhibit thegreatest contractile performance during the animal wake vssleep time, in Langendorff analysis.20 Conversely, this vari-ation in performance is ablated in hearts of diurnal-disruptedmice.81 At a molecular level, day/night rhythms in cardiacmyofilament activity are lost in cardiomyocyte-specific Clockmutant mice compared with wild type mice, thus furthersupporting the notion that cardiac contractile performancelinks to the circadian mechanism.81 That there are signifi-cant disruptions in sarcomere architecture in Bmal1-/- miceprovides further support for this notion.115 Our group alsodemonstrated a functional correlation between myofilamentactivity and protein phosphorylation as one mechanism forregulating diurnal myofilament function in the murineheart.81 Most recently we demonstrated that the expressionof murine cardiac Titin-cap (Tcap, telethonin), a key regu-lator of sarcomere structure and function, is regulated by thecircadian mechanism transcription factors CLOCK andBMAL1.153 In a similar light, Andrews and colleagues haveshown links between skeletal muscles and the circadian clockmechanism.154 Thus collectively, these studies indicate thatthe circadian mechanism plays an important and previouslyunrecognized role in contractile muscle composition andperformance.

Circadian rhythms and myofilament signalling

Cyclic adenosine monophosphate-dependent protein ki-nase (PKA) targets the myofilament complex (troponin I,myosin binding protein C, titin, and others) and thus has a

powerful ability to affect myofilament function. PKA geneexpression is regulated by the cardiomyocyte clock,13 whichmight help explain diurnal variations in myocardial contrac-tility. Additionally, there is diurnal variation in b-adrenergicreceptor activation, a transmembrane receptor coupled withPKA activation in cardiac myocytes.155,156 Diurnal disruptiondecreases myofilament protein phosphorylation by PKA afterMI in mice.103 Together, these findings show the importanceof circadian variations in the b-adrenergic-PKA cascade, formyofilament regulation partly under control of the car-diomyocyte clock mechanism, and a pathway by whichchanges in myofilament function can influence the ability ofthe heart to respond to changes in driving factors.

Circadian mechanism and cardiac electrophysiology

As alluded to herein, the circadian mechanism plays a rolein cardiac contractility via time of day fluctuations in con-tractile proteins. Mechanistically, this might also be regulatedintrinsically by ion channels in the heart that influencemyofilament activation,156,157 some of which exhibit diurnalvariation. Time of day-dependent oscillations have been re-ported in the heart for the voltage-dependent potassiumchannels Kv1.5 and Kv4.2,

158 and the L-type voltage-gatedcalcium channel subunit VGCCa1D.159 Also, L-type cal-cium channel current densities increase during the activeperiod, which is consistent with the nocturnal elevation inmurine HR.155,157 Moreover, circadian variations in potas-sium channel interacting protein-2 affect potassium channelopening in a manner that is consistent with changes in action

Page 7: Consequences of Circadian and Sleep Disturbances for the

866 Canadian Journal of CardiologyVolume 31 2015

potential duration and circadian variations in HR. In vivo, thekruppel-like factor 15 (circadian output gene) knockout micedisplay increased susceptibility to ventricular arrhythmias, andreduced expression of potassium channel interacting protein2.160 Finally, there is loss of the rhythmic sodium channelvoltage-gated type V a subunit, and increased susceptibility toarrhythmias, in inducible cardiomyocyte-specific Bmal1-/-

mice.161 These studies provide new insights for understandingthe diurnal nature of cardiac electrophysiology, HR regula-tion, and arrhythmias.

New Frontiers: Translational ChronocardiologyAnother exciting new frontier is knowledge translation.

This is important for rhythms research to have a beneficialeffect on the quality of life for people with heart disease. Onearea of application is chronobiomarkers, in which temporalgene (microarray) and protein (proteomic) metadata are usedto identify new biomarkers of heart disease. A second area ofapplication is chronotherapy, which involves timing therapiesto improve the management of cardiovascular (and other)diseases. These approaches and relevance to clinical cardiologyare described in more detail herein.

Chronobiomarkers

Applying circadian concepts to clinical cardiology offersnew opportunities for discovering biomarkers, with a varietyof different technical approaches. For example, Ueda et al.estimate tissue time of day in mice by measuring expression ofapproximately 100 genes across a molecular time table inmicroarray analyses.162 Body time in mice163 and humans164

can also be by estimated using metabolomic studies. Wediscovered time of day biomarkers in murine cardiac hyper-trophy using microarray profiling and a novel time seriesalgorithm termed DeltaGene,165 and de novo chrono-biomarkers in murine plasma samples over 24-hour pe-riods.166 Collectively, these preclinical proof of conceptstudies are important for designing new approaches forbiomarker discovery, by taking advantage of the body’s nat-ural 24-hour molecular rhythms. Moreover, they have prac-tical application for chronotherapy (described herein), whichrequires easily accessible markers of body time to optimize thetiming of drug treatments.

Chronotherapy benefits (reverse) cardiac remodelling

Chronotherapy improves therapeutic efficacy (and possiblyreduces toxicity) by timing treatments with our daily physi-ology. For example, in a preclinical study on heart disease, weshowed that sleep-time administration of the short-actingangiotensin-converting enzyme inhibitor captopril markedlyreduced cardiac remodelling in mice with pressure overload-induced cardiac hypertrophy; conversely, administration atwake time had no measureable benefit on heart damage.167

Mechanistically, the sleep-time benefit is associated withdiurnal rhythms in the renin-angiotensin-aldosterone systemand cardiac genes and proteins, which peak during the rodentsleep time. Indeed, there is significant potential for chrono-therapy, because it has been recently reported that many best-selling drugs and the World Health Organization essential

medicines have short half-lives for directly targeting genepathways at specific times of day or night.168

Hypertension chronotherapy

Chronotherapy can also be effective in the treatment ofhypertension. Most people have a diurnal BP profile thatexhibits a dip of approximately 10% at night.18 Conversely,hypertensive nondippers169 or normotensive individuals withattenuated nocturnal dips in BP170 have an increased risk ofheart disease. Ambulatory BP monitoring, and especiallycovering the night time, is a useful prognostic indicator forheart disease,171-173 and is recommended by many interna-tional societies.174 Chronotherapy to treat hypertension atnight helps to restore the day/night BP rhythms and reducesthe risk of cardiovascular disease.175-178 For example, dayand night drug administration manage daytime hyperten-sion, however, only nighttime administration of ramipril,179

or telmisartan,180 or valsartan and amlodipine181 restores thenocturnal BP dip. Furthermore, the clinical trial termedAmbulatory Blood Pressure Monitoring in the Prediction ofCardiovascular Events and Effects of Chronotherapy(MAPEC) demonstrated prospectively that bedtime chro-notherapy but not morning administration of antihyper-tensive medication improves BP control and reducescardiovascular disease risk.182

Chronotherapy and other cardiovascular conditions:obstructive sleep apnea, hemodialysis, aspirin

Chronotherapy also holds significant clinical applicabilityin other conditions associated with heart disease. For example,obstructive sleep apnea (OSA) is a common respiratory dis-order of sleep, with underlying circadian-coupled mechanismsthat if left untreated can lead to the development and exac-erbation of heart disease and heart failure.183-188 Conversely,nocturnal continuous positive airway pressure (CPAP) therapybenefits patients with OSA and heart failure, reduces sym-pathetic activity,189,190 decreases BP during sleep, and im-proves left ventricular function.191-194 At a molecularcircadian level OSA appears to cause circadian clock genedysfunction, and nocturnal CPAP therapy helps to improve it.This was demonstrated by comparing Per1 mRNA expressionin peripheral blood cells of healthy subjects, OSA patients,and OSA patients treated with CPAP.195

Several additional lines of evidence further support thenotion that timing of therapy benefits the cardiovascularsystem. For example, nocturnal hemodialysis of patients withcardio-renal disease reduces left ventricular hypertrophy196,197

and improves myocardial function,198 compared with patientsgiven conventional daytime hemodialysis. In another study,low-dose aspirin taken at night reduced the circadian rhythmof platelet reactivity in healthy individuals; it is postulated thatthis might also reduce the risk of acute cardiovascular eventsduring the peak morning hours.199 Thus, these time-dependent effects of therapies have significant potential forbenefitting many patients in clinical settings.

Conclusions and Final DirectionsClinical and experimental studies demonstrate the impor-

tant role of circadian rhythms for healthy cardiovascular

Page 8: Consequences of Circadian and Sleep Disturbances for the

Alibhai et al. 867Circadian Rhythm Disturbance and Heart Disease

physiology. Disturbing of rhythms has direct adverse conse-quences on cardiac physiology and pathophysiology. Futurestudies will undoubtedly provide new insights into themechanisms and molecular pathways that drive these pathol-ogies. In terms of translation, significant opportunities exist toincrease our understanding of circadian rhythms on cardio-vascular disease. For example, experiments investigatingcircadian regulation of cerebrovascular conditions and strokeare virtually unknown despite this being the third leadingcause of death in Canada with an estimated 50,000 strokes peryear.200 Also, metabolic disorders are considered major riskfactors for cardiovascular disease, and thus circadian rhythmsresearch applied to prevalent clinical conditions such as dia-betes and obesity could be fruitful areas of investigation.Determining the relationship between the circadian mecha-nism and cardiopulmonary conditions (eg, OSA and others)provides new opportunities to improve the diagnosis andtreatment of heart and lung disease. Upcoming areas forknowledge translation include development of databases forpatients with heart disease concurrent with circadian and/orsleep disorders, collecting circadian biometric data (eg, 24-hour HR and BP profiles, gene and protein biomarkers) inchronobiology and sleep clinics, and chronotherapeutic stra-tegies to improve the effectiveness of existing treatments.Thus, circadian rhythms research advances our understandingof cardiovascular health and disease, and leads to novel stra-tegies for management and treatment of heart disease inclinical settings.

Funding SourcesCanadian Institutes of Health Research grant to T.A.

Martino.

DisclosuresThe authors have no conflicts of interest to disclose.

References

1. Aschoff J. Circadian rhythms in man. Science 1965;148:1427-32.

2. Hastings MH, Reddy AB, Maywood ES. A clockwork web: circadiantiming in brain and periphery, in health and disease. Nat Rev Neurosci2003;4:649-61.

3. Reppert SM, Weaver DR. Coordination of circadian timing in mam-mals. Nature 2002;418:935-41.

4. Roenneberg T, Merrow M. Circadian clocks - the fall and rise ofphysiology. Nat Rev Mol Cell Biol 2005;6:965-71.

5. Durgan DJ, Young ME. The cardiomyocyte circadian clock: emergingroles in health and disease. Circ Res 2010;106:647-58.

6. Martino TA, Sole MJ. Molecular time: an often overlooked dimensionto cardiovascular disease. Circ Res 2009;105:1047-61.

7. Paschos GK, FitzGerald GA. Circadian clocks and vascular function.Circ Res 2010;106:833-41.

8. Ko CH, Takahashi JS. Molecular components of the mammaliancircadian clock. Hum Mol Genet 2006;15:R271-7.

9. Dardente H, Cermakian N. Molecular circadian rhythms in central andperipheral clocks in mammals. Chronobiol Int 2007;24:195-213.

10. Gamble KL, Berry R, Frank SJ, Young ME. Circadian clock control ofendocrine factors. Nat Rev Endocrinol 2014;10:466-75.

11. Evans JA, Davidson AJ. Health consequences of circadian disruption inhumans and animal models. Prog Mol Biol Transl Sci 2013;119:283-323.

12. Sahar S, Sassone-Corsi P. Metabolism and cancer: the circadian clockconnection. Nat Rev Cancer 2009;9:886-96.

13. Bray MS, Shaw CA, Moore MW, et al. Disruption of the circadianclock within the cardiomyocyte influences myocardial contractilefunction, metabolism, and gene expression. Am J Physiol Heart CircPhysiol 2008;294:H1036-47.

14. Boivin DB. Influence of sleep-wake and circadian rhythm disturbancesin psychiatric disorders. J Psychiatry Neurosci 2000;25:446-58.

15. Smolensky MH, Portaluppi F, Manfredini R, et al. Diurnal and twenty-four hour patterning of human diseases: cardiac, vascular, and respira-tory diseases, conditions, and syndromes. Sleep Med Rev 2014;14:1-9.

16. Clarke JM, Hamer J, Shelton JR, Taylor S, Venning GR. The rhythmof the normal human heart. Lancet 1976;1:508-12.

17. Floras JS, Jones JV, Johnston JA, et al. Arousal and the circadian rhythmof blood pressure. Clin Sci Mol Med Suppl 1978;4:395s-7s.

18. Millar-Craig MW, Bishop CN, Raftery EB. Circadian variation ofblood-pressure. Lancet 1978;1:795-7.

19. Scheer FA, Hu K, Evoniuk H, et al. Impact of the human circadiansystem, exercise, and their interaction on cardiovascular function. ProcNatl Acad Sci U S A 2010;107:20541-6.

20. Young ME, Razeghi P, Cedars AM, Guthrie PH, Taegtmeyer H.Intrinsic diurnal variations in cardiac metabolism and contractile func-tion. Circ Res 2001;89:1199-208.

21. Saleh MA, Winget CM. Effect of suprachiasmatic lesions on diurnalheart rate rhythm in the rat. Physiol Behav 1977;19:561-4.

22. Warren WS, Champney TH, Cassone VM. The suprachiasmatic nu-cleus controls the circadian rhythm of heart rate via the sympatheticnervous system. Physiol Behav 1994;55:1091-9.

23. Janssen BJ, Tyssen CM, Duindam H, Rietveld WJ. Suprachiasmaticlesions eliminate 24-h blood pressure variability in rats. Physiol Behav1994;55:307-11.

24. Takezawa H, Hayashi H, Sano H, Saito H, Ebihara S. Circadian andestrous cycle-dependent variations in blood pressure and heart rate infemale rats. Am J Physiol 1994;267:R1250-6.

25. Sei H, Oishi K, Chikahisa S, et al. Diurnal amplitudes of arterialpressure and heart rate are dampened in Clock mutant mice andadrenalectomized mice. Endocrinology 2008;149:3576-80.

26. Curtis AM, Cheng Y, Kapoor S, et al. Circadian variation of bloodpressure and the vascular response to asynchronous stress. Proc NatlAcad Sci U S A 2007;104:3450-5.

27. Muller JE, Stone PH, Turi ZG, et al. Circadian variation in the fre-quency of onset of acute myocardial infarction. N Engl J Med1985;313:1315-22.

28. Cohen MC, Rohtla KM, Lavery CE, Muller JE, Mittleman MA. Meta-analysis of the morning excess of acute myocardial infarction and suddencardiac death. Am J Cardiol 1997;79:1512-6.

29. Kanth R, Ittaman S, Rezkalla S. Circadian patterns of ST elevationmyocardial infarction in the new millennium. Clin Med Res 2013;11:66-72.

Page 9: Consequences of Circadian and Sleep Disturbances for the

868 Canadian Journal of CardiologyVolume 31 2015

30. Willich SN, Lowel H, Lewis M, et al. Association of wake time and theonset of myocardial infarction. Triggers and mechanisms of myocardialinfarction (TRIMM) pilot study. TRIMM Study Group. Circulation1991;84:VI62-7.

31. Tofler GH, Muller JE, Stone PH, et al. Modifiers of timing and possibletriggers of acute myocardial infarction in the Thrombolysis inMyocardial Infarction Phase II (TIMI II) Study Group. J Am CollCardiol 1992;20:1049-55.

32. Tofler GH, Stone PH, Maclure M, et al. Analysis of possible triggers ofacute myocardial infarction (the MILIS study). Am J Cardiol 1990;66:22-7.

33. Goldberg RJ, Brady P, Muller JE, et al. Time of onset of symptoms ofacute myocardial infarction. Am J Cardiol 1990;66:140-4.

34. Durgan DJ, Pulinilkunnil T, Villegas-Montoya C, et al. Shortcommunication: ischemia/reperfusion tolerance is time-of-day-dependent: mediation by the cardiomyocyte circadian clock. Circ Res2010;106:546-50.

35. Reiter R, Swingen C, Moore L, Henry TD, Traverse JH. Circadiandependence of infarct size and left ventricular function after ST eleva-tion myocardial infarction. Circ Res 2012;110:105-10.

36. Suarez-Barrientos A, Lopez-Romero P, Vivas D, et al. Circadian varia-tions of infarct size in acute myocardial infarction. Heart 2011;97:970-6.

37. Marler JR, Price TR, Clark GL, et al. Morning increase in onset ofischemic stroke. Stroke 1989;20:473-6.

38. Cannon CP, McCabe CH, Stone PH, et al. Circadian variation in theonset of unstable angina and non-Q-wave acute myocardial infarction(the TIMI III Registry and TIMI IIIB). Am J Cardiol 1997;79:253-8.

39. Tofler GH, Gebara OC, Mittleman MA, et al. Morning peak in ven-tricular tachyarrhythmias detected by time of implantable cardioverter/defibrillator therapy. The CPI Investigators. Circulation 1995;92:1203-8.

40. Behrens S, Galecka M, Bruggemann T, et al. Circadian variation ofsustained ventricular tachyarrhythmias terminated by appropriate shocksin patients with an implantable cardioverter defibrillator. Am Heart J1995;130:79-84.

41. Eksik A, Akyol A, Norgaz T, et al. Circadian pattern of spontaneousventricular tachyarrhythmias in patients with implantable cardioverterdefibrillators. Med Sci Monit 2007;13:CR412-6.

42. Venditti FJ Jr, John RM, Hull M, et al. Circadian variation in defi-brillation energy requirements. Circulation 1996;94:1607-12.

43. Kong TQ Jr, Goldberger JJ, Parker M, Wang T, Kadish AH. Circadianvariation in human ventricular refractoriness. Circulation 1995;92:1507-16.

44. Willich SN, Levy D, Rocco MB, et al. Circadian variation in theincidence of sudden cardiac death in the Framingham Heart Studypopulation. Am J Cardiol 1987;60:801-6.

45. Muller JE, Ludmer PL, Willich SN, et al. Circadian variation in thefrequency of sudden cardiac death. Circulation 1987;75:131-8.

46. Willich SN, Goldberg RJ, Maclure M, Perriello L, Muller JE. Increasedonset of sudden cardiac death in the first three hours after awakening.Am J Cardiol 1992;70:65-8.

47. Fava S, Azzopardi J, Muscat HA, Fenech FF. Absence of circadianvariation in the onset of acute myocardial infarction in diabetic subjects.Br Heart J 1995;74:370-2.

48. Kuniyoshi FH, Garcia-Touchard A, Gami AS, et al. Day-night variationof acute myocardial infarction in obstructive sleep apnea. J Am CollCardiol 2008;52:343-6.

49. Zarich S, Waxman S, Freeman RT, et al. Effect of autonomic nervoussystem dysfunction on the circadian pattern of myocardial ischemia indiabetes mellitus. J Am Coll Cardiol 1994;24:956-62.

50. Ridker PM, Manson JE, Buring JE, Muller JE, Hennekens CH.Circadian variation of acute myocardial infarction and the effect of low-dose aspirin in a randomized trial of physicians. Circulation 1990;82:897-902.

51. Ruggeri ZM. Platelets in atherothrombosis. Nat Med 2002;8:1227-34.

52. Scheer FA, Michelson AD, Frelinger AL 3rd, et al. The humanendogenous circadian system causes greatest platelet activation duringthe biological morning independent of behaviors. PLoS One 2011;6:e24549.

53. Tofler GH, Brezinski D, Schafer AI, et al. Concurrent morning increasein platelet aggregability and the risk of myocardial infarction and suddencardiac death. N Engl J Med 1987;316:1514-8.

54. Andrews NP, Gralnick HR, Merryman P, Vail M, Quyyumi AA.Mechanisms underlying the morning increase in platelet aggregation: aflow cytometry study. J Am Coll Cardiol 1996;28:1789-95.

55. Decousus HA, Croze M, Levi FA, et al. Circadian changes in antico-agulant effect of heparin infused at a constant rate. Br Med J (Clin ResEd) 1985;290:341-4.

56. Scheer FA, Shea SA. Human circadian system causes a morning peak inprothrombotic plasminogen activator inhibitor-1 (PAI-1) independentof the sleep/wake cycle. Blood 2014;123:590-3.

57. Otto ME, Svatikova A, Barretto RB, et al. Early morning attenuation ofendothelial function in healthy humans. Circulation 2004;109:2507-10.

58. Hu K, Ivanov P, Hilton MF, et al. Endogenous circadian rhythm in anindex of cardiac vulnerability independent of changes in behavior. ProcNatl Acad Sci U S A 2004;101:18223-7.

59. Maemura K, de la Monte SM, Chin MT, et al. CLIF, a novel cycle-likefactor, regulates the circadian oscillation of plasminogen activatorinhibitor-1 gene expression. J Biol Chem 2000;275:36847-51.

60. Schoenhard JA, Smith LH, Painter CA, et al. Regulation of the PAI-1promoter by circadian clock components: differential activation byBMAL1 and BMAL2. J Mol Cell Cardiol 2003;35:473-81.

61. Oishi K, Miyazaki K, Uchida D, et al. PERIOD2 is a circadian negativeregulator of PAI-1 gene expression in mice. J Mol Cell Cardiol 2009;46:545-52.

62. Durgan DJ, Moore MW, Ha NP, et al. Circadian rhythms inmyocardial metabolism and contractile function: influence of workloadand oleate. Am J Physiol Heart Circ Physiol 2007;293:H2385-93.

63. Durgan DJ, Pat BM, Laczy B, et al. O-GlcNAcylation, novel post-translational modification linking myocardial metabolism and car-diomyocyte circadian clock. J Biol Chem 2011;286:44606-19.

64. Chatham JC, Young ME. Regulation of myocardial metabolism by thecardiomyocyte circadian clock. J Mol Cell Cardiol 2013;55:139-46.

65. Tsai JY, Kienesberger PC, Pulinilkunnil T, et al. Direct regulation ofmyocardial triglyceride metabolism by the cardiomyocyte circadianclock. J Biol Chem 2010;285:2918-29.

66. Young ME, Brewer RA, Peliciari-Garcia RA, et al. Cardiomyocyte-specific BMAL1 plays critical roles in metabolism, signaling, and

Page 10: Consequences of Circadian and Sleep Disturbances for the

Alibhai et al. 869Circadian Rhythm Disturbance and Heart Disease

maintenance of contractile function of the heart. J Biol Rhythms2014;29:257-76.

67. Storch KF, Lipan O, Leykin I, et al. Extensive and divergent circadiangene expression in liver and heart. Nature 2002;417:78-83.

68. Martino T, Arab S, Straume M, et al. Day/night rhythms in geneexpression of the normal murine heart. J Mol Med (Berl) 2004;82:256-64.

69. Young ME, Razeghi P, Taegtmeyer H. Clock genes in the heart:characterization and attenuation with hypertrophy. Circ Res 2001;88:1142-50.

70. de Lichtenberg U, Jensen LJ, Fausboll A, et al. Comparison ofcomputational methods for the identification of cell cycle-regulatedgenes. Bioinformatics 2005;21:1164-71.

71. Glynn EF, Chen J, Mushegian AR. Detecting periodic patterns inunevenly spaced gene expression time series using Lomb-Scargleperiodograms. Bioinformatics 2006;22:310-6.

72. Hughes ME, Hogenesch JB, Kornacker K. JTK_CYCLE: an efficientnonparametric algorithm for detecting rhythmic components ingenome-scale data sets. J Biol Rhythms 2010;25:372-80.

73. Pizarro A, Hayer K, Lahens NF, Hogenesch JB. CircaDB: a database ofmammalian circadian gene expression profiles. Nucleic Acids Res2013;41:D1009-13.

74. Leibetseder V, Humpeler S, Svoboda M, et al. Clock genes displayrhythmic expression in human hearts. Chronobiol Int 2009;26:621-36.

75. Davidson AJ, London B, Block GD, Menaker M. Cardiovascular tissuescontain independent circadian clocks. Clin Exp Hypertens 2005;27:307-11.

76. Balsalobre A, Damiola F, Schibler U. A serum shock induces circadiangene expression in mammalian tissue culture cells. Cell 1998;93:929-37.

77. Durgan DJ, Hotze MA, Tomlin TM, et al. The intrinsic circadian clockwithin the cardiomyocyte. Am J Physiol Heart Circ Physiol 2005;289:H1530-41.

78. McNamara P, Seo SB, Rudic RD, et al. Regulation of CLOCK andMOP4 by nuclear hormone receptors in the vasculature: a humoralmechanism to reset a peripheral clock. Cell 2001;105:877-89.

79. Chalmers JA, Lin SY, Martino TA, et al. Diurnal profiling of neuro-endocrine genes in murine heart, and shift in proopiomelanocortin geneexpression with pressure-overload cardiac hypertrophy. J Mol Endo-crinol 2008;41:117-24.

80. Chalmers JA, Martino TA, Tata N, et al. Vascular circadian rhythms ina mouse vascular smooth muscle cell line (Movas-1). Am J Physiol RegulIntegr Comp Physiol 2008;295:R1529-38.

81. Podobed P, Pyle WG, Ackloo S, et al. The day/night proteome in themurine heart. Am J Physiol Regul Integr Comp Physiol 2014;307:R121-37.

82. Reddy AB, Karp NA, Maywood ES, et al. Circadian orchestration of thehepatic proteome. Curr Biol 2006;16:1107-15.

83. Luck S, Thurley K, Thaben PF, Westermark PO. Rhythmic degrada-tion explains and unifies circadian transcriptome and proteome data.Cell Rep 2014;9:741-51.

84. Ishida A, Mutoh T, Ueyama T, et al. Light activates the adrenal gland:timing of gene expression and glucocorticoid release. Cell Metab2005;2:297-307.

85. Lutzner N, Kalbacher H, Krones-Herzig A, Rosl F. FOXO3 is aglucocorticoid receptor target and regulates LKB1 and its own

expression based on cellular AMP levels via a positive autoregulatoryloop. PLoS One 2012;7:e42166.

86. Curtis AM, Seo SB, Westgate EJ, et al. Histone acetyltransferase-dependent chromatin remodeling and the vascular clock. J Biol Chem2004;279:7091-7.

87. Innominato PF, Levi FA, Bjarnason GA. Chronotherapy and the mo-lecular clock: clinical implications in oncology. Adv Drug Deliv Rev2010;62:979-1001.

88. Sole MJ, Martino TA. Diurnal physiology: core principles with appli-cation to the pathogenesis, diagnosis, prevention, and treatment ofmyocardial hypertrophy and failure. J Appl Physiol 2009;107:1318-27.

89. Bass J, Takahashi JS. Circadian integration of metabolism and ener-getics. Science 2010;330:1349-54.

90. Frangogiannis NG, Smith CW, Entman ML. The inflammatoryresponse in myocardial infarction. Cardiovasc Res 2002;53:31-47.

91. Mann DL. The emerging role of innate immunity in the heart andvascular system: for whom the cell tolls. Circ Res 2011;108:1133-45.

92. Nian M, Lee P, Khaper N, Liu P. Inflammatory cytokines and post-myocardial infarction remodeling. Circ Res 2004;94:1543-53.

93. Abo T, Kawate T, Itoh K, Kumagai K. Studies on the bioperiodicity ofthe immune response. I. Circadian rhythms of human T, B, and K celltraffic in the peripheral blood. J Immunol 1981;126:1360-3.

94. Born J, Lange T, Hansen K, Molle M, Fehm HL. Effects of sleep andcircadian rhythm on human circulating immune cells. J Immunol1997;158:4454-64.

95. Haus E, Lakatua DJ, Swoyer J, Sackett-Lundeen L. Chronobiology inhematology and immunology. Am J Anat 1983;168:467-517.

96. Haus E, Smolensky MH. Biologic rhythms in the immune system.Chronobiol Int 1999;16:581-622.

97. Fortier EE, Rooney J, Dardente H, et al. Circadian variation of theresponse of T cells to antigen. J Immunol 2011;187:6291-300.

98. Boivin DB, James FO, Wu A, et al. Circadian clock genes oscillate inhuman peripheral blood mononuclear cells. Blood 2003;102:4143-5.

99. Bollinger T, Leutz A, Leliavski A, et al. Circadian clocks in mouse andhuman CD4þ T cells. PLoS One 2011;6:e29801.

100. Oishi K, Ohkura N, Kadota K, et al. Clock mutation affects circadianregulation of circulating blood cells. J Circadian Rhythms 2006;4:13.

101. Cermakian N, Lange T, Golombek D, et al. Crosstalk between thecircadian clock circuitry and the immune system. Chronobiol Int2013;30:870-88.

102. Keller M, Mazuch J, Abraham U, et al. A circadian clock in macro-phages controls inflammatory immune responses. Proc Natl Acad SciU S A 2009;106:21407-12.

103. Alibhai FJ, Tsimakouridze EV, Chinnappareddy N, et al. Short-termdisruption of diurnal rhythms after murine myocardial infarctionadversely affects long-term myocardial structure and function. Circ Res2014;114:1713-22.

104. Patel M, Chipman J, Carlin BW, Shade D. Sleep in the intensive careunit setting. Crit Care Nurs Q 2008;31:309-18.

105. Buxton OM, Ellenbogen JM, Wang W, et al. Sleep disruption due tohospital noises: a prospective evaluation. Ann Intern Med 2012;157:170-9.

106. Drouot X, Cabello B, d’Ortho MP, Brochard L. Sleep in the intensivecare unit. Sleep Med Rev 2008;12:391-403.

Page 11: Consequences of Circadian and Sleep Disturbances for the

870 Canadian Journal of CardiologyVolume 31 2015

107. Chan MC, Spieth PM, Quinn K, et al. Circadian rhythms: from basicmechanisms to the intensive care unit. Crit Care Med 2012;40:246-53.

108. Stecker EC. How can I recover if you won’t let me sleep? Sci TranslMed 2014;6:231ec65.

109. Penev PD, Kolker DE, Zee PC, Turek FW. Chronic circadiandesynchronization decreases the survival of animals with car-diomyopathic heart disease. Am J Physiol 1998;275:H2334-7.

110. Martino TA, Tata N, Belsham DD, et al. Disturbed diurnal rhythmalters gene expression and exacerbates cardiovascular disease with rescueby resynchronization. Hypertension 2007;49:1104-13.

111. Lowrey PL, Shimomura K, Antoch MP, et al. Positional synteniccloning and functional characterization of the mammalian circadianmutation tau. Science 2000;288:483-92.

112. Ralph MR, Menaker M. A mutation of the circadian system in goldenhamsters. Science 1988;241:1225-7.

113. Hurd MW, Ralph MR. The significance of circadian organization forlongevity in the golden hamster. J Biol Rhythms 1998;13:430-6.

114. Martino TA, Oudit GY, Herzenberg AM, et al. Circadian rhythmdisorganization produces profound cardiovascular and renal disease inhamsters. Am J Physiol Regul Integr Comp Physiol 2008;294:R1675-83.

115. Lefta M, Campbell KS, Feng HZ, Jin JP, Esser KA. Development ofdilated cardiomyopathy in Bmal1-deficient mice. Am J Physiol HeartCirc Physiol 2012;303:H475-85.

116. Durgan DJ, Tsai JY, Grenett MH, et al. Evidence suggesting that thecardiomyocyte circadian clock modulates responsiveness of the heart tohypertrophic stimuli in mice. Chronobiol Int 2011;28:187-203.

117. Woon PY, Kaisaki PJ, Braganca J, et al. Aryl hydrocarbon receptornuclear translocator-like (BMAL1) is associated with susceptibility tohypertension and type 2 diabetes. Proc Natl Acad Sci U S A 2007;104:14412-7.

118. Englund A, Kovanen L, Saarikoski ST, et al. NPAS2 and PER2 arelinked to risk factors of the metabolic syndrome. J Circadian Rhythms2009;7:5.

119. Wang Q, Maillard M, Schibler U, Burnier M, Gachon F. Cardiachypertrophy, low blood pressure, and low aldosterone levels in micedevoid of the three circadian PAR bZip transcription factors DBP, HLF,and TEF. Am J Physiol Regul Integr Comp Physiol 2010;299:R1013-9.

120. Durgan DJ, Trexler NA, Egbejimi O, et al. The circadian clock withinthe cardiomyocyte is essential for responsiveness of the heart to fattyacids. J Biol Chem 2006;281:24254-69.

121. Virag JA, Dries JL, Easton PR, et al. Attenuation of myocardial injury inmice with functional deletion of the circadian rhythm gene mPer2. AmJ Physiol Heart Circ Physiol 2010;298:H1088-95.

122. Eckle T, Hartmann K, Bonney S, et al. Adora2b-elicited Per2 stabili-zation promotes a HIF-dependent metabolic switch crucial formyocardial adaptation to ischemia. Nat Med 2012;18:774-82.

123. World Health Organization. World Health Report 2002: ReducingRisks, Promoting Healthy Life. Available at: http://www.who.int/whr/2002/en/. Accessed August 10, 2014.

124. Furlan R, Barbic F, Piazza S, et al. Modifications of cardiac autonomicprofile associated with a shift schedule of work. Circulation 2000;102:1912-6.

125. Chau NP, Mallion JM, de Gaudemaris R, et al. Twenty-four-hourambulatory blood pressure in shift workers. Circulation 1989;80:341-7.

126. Scheer FA, Hilton MF, Mantzoros CS, Shea SA. Adverse metabolic andcardiovascular consequences of circadian misalignment. Proc Natl AcadSci U S A 2009;106:4453-8.

127. Knutsson A, Akerstedt T, Jonsson BG, Orth-Gomer K. Increased risk ofischaemic heart disease in shift workers. Lancet 1986;2:89-92.

128. Kawachi I, Colditz GA, Stampfer MJ, et al. Prospective study of shiftwork and risk of coronary heart disease in women. Circulation 1995;92:3178-82.

129. Vyas MV, Garg AX, Iansavichus AV, et al. Shift work and vascularevents: systematic review and meta-analysis. BMJ 2012;345:e4800.

130. Haupt CM, Alte D, Dorr M, et al. The relation of exposure to shiftwork with atherosclerosis and myocardial infarction in a general pop-ulation. Atherosclerosis 2008;201:205-11.

131. Oishi M, Suwazono Y, Sakata K, et al. A longitudinal study on therelationship between shift work and the progression of hypertension inmale Japanese workers. J Hypertens 2005;23:2173-8.

132. Centers for Disease Control and Prevention. WorkSchedules: ShiftWork and Long Work Hours. Available at: http://www.cdc.gov/niosh/topics/workschedules/. Accessed August 10, 2014.

133. Carrier J, Monk TH. Circadian rhythms of performance: new trends.Chronobiol Int 2000;17:719-32.

134. Toh KL, Jones CR, He Y, et al. An hPer2 phosphorylation site mu-tation in familial advanced sleep phase syndrome. Science 2001;291:1040-3.

135. National Sleep Foundation. 2005 Sleep in America Poll. Available at:http://sleepfoundation.org/sites/default/files/2005_summary_of_findings.pdf. Accessed August 10, 2014.

136. Ayas NT, White DP, Manson JE, et al. A prospective study of sleepduration and coronary heart disease in women. Arch Intern Med2003;163:205-9.

137. Wingard DL, Berkman LF. Mortality risk associated with sleepingpatterns among adults. Sleep 1983;6:102-7.

138. Kripke DF, Simons RN, Garfinkel L, Hammond EC. Short and longsleep and sleeping pills. Is increased mortality associated? Arch GenPsychiatry 1979;36:103-16.

139. Healthy Sleep. Sleep and Disease Risk. Available at: http://healthysleep.med.harvard.edu/healthy/matters/consequences/sleep-and-disease-risk.Accessed on August 10, 2014.

140. Meerlo P, Sgoifo A, Suchecki D. Restricted and disrupted sleep: effectson autonomic function, neuroendocrine stress systems and stressresponsivity. Sleep Med Rev 2008;12:197-210.

141. Buxton OM, Cain SW, O’Connor SP, et al. Adverse metabolic con-sequences in humans of prolonged sleep restriction combined withcircadian disruption. Sci Transl Med 2012;4:129ra43.

142. Anafi RC, Pellegrino R, Shockley KR, et al. Sleep is not just for thebrain: transcriptional responses to sleep in peripheral tissues. BMCGenomics 2013;14:362.

143. Duffy JF, Dijk DJ, Hall EF, Czeisler CA. Relationship of endogenouscircadian melatonin and temperature rhythms to self-reported prefer-ence for morning or evening activity in young and older people.J Investig Med 1999;47:141-50.

144. Merikanto I, Lahti T, Puolijoki H, et al. Associations of chronotype andsleep with cardiovascular diseases and type 2 diabetes. Chronobiol Int2013;30:470-7.

Page 12: Consequences of Circadian and Sleep Disturbances for the

Alibhai et al. 871Circadian Rhythm Disturbance and Heart Disease

145. Wittmann M, Dinich J, Merrow M, Roenneberg T. Social jetlag:misalignment of biological and social time. Chronobiol Int 2006;23:497-509.

146. Rutters F, Lemmens SG, Adam TC, et al. Is social jetlag associated withan adverse endocrine, behavioral, and cardiovascular risk profile? J BiolRhythms 2014;29:377-83.

147. Czeisler CA. Perspective: casting light on sleep deficiency. Nature2013;497:S13.

148. Chang A, Aeschbach D, Duffy JF, Czeisler CA. Evening use of light-emitting eReaders negatively affects sleep, circadian timing, and next-morning alertness. Proc Natl Acad Sci U S A 2015;112:1232-7.

149. Pyle WG, Solaro RJ. At the crossroads of myocardial signaling: the roleof Z-discs in intracellular signaling and cardiac function. Circ Res2004;94:296-305.

150. Frank D, Frey N. Cardiac Z-disc signaling network. J Biol Chem2011;286:9897-904.

151. Solaro RJ, Kobayashi T. Protein phosphorylation and signal trans-duction in cardiac thin filaments. J Biol Chem 2011;286:9935-40.

152. Palmer BM. Thick filament proteins and performance in human heartfailure. Heart Fail Rev 2005;10:187-97.

153. Podobed PS, Alibhai FJ, Chow CW, Martino TA. Circadian regulationof myocardial sarcomeric Titin-cap (Tcap, Telethonin): identification ofcardiac clock-controlled genes using open access bioinformatics data.PLoS One 2014;9:e104907.

154. Andrews JL, Zhang X, McCarthy JJ, et al. CLOCK and BMAL1regulate MyoD and are necessary for maintenance of skeletal musclephenotype and function. Proc Natl Acad Sci U S A 2010;107:19090-5.

155. Collins HE, Rodrigo GC. Inotropic response of cardiac ventricularmyocytes to beta-adrenergic stimulation with isoproterenol exhibitsdiurnal variation: involvement of nitric oxide. Circ Res 2010;106:1244-52.

156. Collins HE, Turrell HE, Samani NJ, Rodrigo GC. Diurnal variation inexcitation-contraction coupling is lost in the adult spontaneously hy-pertensive rat heart. J Hypertens 2013;31:1214-23.

157. Ko ML, Shi L, Grushin K, Nigussie F, Ko GY. Circadian profiles in theembryonic chick heart: L-type voltage-gated calcium channels andsignaling pathways. Chronobiol Int 2010;27:1673-96.

158. Yamashita T, Sekiguchi A, Iwasaki YK, et al. Circadian variation ofcardiac Kþ channel gene expression. Circulation 2003;107:1917-22.

159. Ko ML, Shi L, Tsai JY, et al. Cardiac-specific mutation of Clock altersthe quantitative measurements of physical activities without changingbehavioral circadian rhythms. J Biol Rhythms 2011;26:412-22.

160. Jeyaraj D, Haldar SM, Wan X, et al. Circadian rhythms govern cardiacrepolarization and arrhythmogenesis. Nature 2012;483:96-9.

161. Schroder EA, Lefta M, Zhang X, et al. The cardiomyocyte molecularclock, regulation of Scn5a, and arrhythmia susceptibility. Am J PhysiolCell Physiol 2013;304:C954-65.

162. Ueda HR, Chen W, Minami Y, et al. Molecular-timetable methods fordetection of body time and rhythm disorders from single-time-pointgenome-wide expression profiles. Proc Natl Acad Sci U S A2004;101:11227-32.

163. Minami Y, Kasukawa T, Kakazu Y, et al. Measurement of internal bodytime by blood metabolomics. Proc Natl Acad Sci U S A 2009;106:9890-5.

164. Kasukawa T, Sugimoto M, Hida A, et al. Human blood metabolitetimetable indicates internal body time. Proc Natl Acad Sci U S A2012;109:15036-41.

165. Tsimakouridze EV, Straume M, Podobed PS, et al. Chronomics ofpressure overload-induced cardiac hypertrophy in mice reveals alteredday/night gene expression and biomarkers of heart disease. ChronobiolInt 2012;29:810-21.

166. Martino TA, Tata N, Bjarnason GA, Straume M, Sole MJ. Diurnalprotein expression in blood revealed by high throughput mass spec-trometry proteomics and implications for translational medicine andbody time of day. Am J Physiol Regul Integr Comp Physiol 2007;293:R1430-7.

167. Martino TA, Tata N, Simpson JA, et al. The primary benefits ofangiotensin-converting enzyme inhibition on cardiac remodeling occurduring sleep time in murine pressure overload hypertrophy. J Am CollCardiol 2011;57:2020-8.

168. Zhang R, Lahens NF, Ballance HI, Hughes ME, Hogenesch JB.A circadian gene expression atlas in mammals: implications for biologyand medicine. Proc Natl Acad Sci U S A 2014;111:16219-24.

169. Verdecchia P, Schillaci G, Gatteschi C, et al. Blunted nocturnal fall inblood pressure in hypertensive women with future cardiovascularmorbid events. Circulation 1993;88:986-92.

170. Ohkubo T, Hozawa A, Yamaguchi J, et al. Prognostic significance of thenocturnal decline in blood pressure in individuals with and without high24-h blood pressure: the Ohasama study. J Hypertens 2002;20:2183-9.

171. Clement DL, De Buyzere ML, De Bacquer DA, et al. Prognostic valueof ambulatory blood-pressure recordings in patients with treated hy-pertension. N Engl J Med 2003;348:2407-15.

172. Perloff D, Sokolow M, Cowan R. The prognostic value of ambulatoryblood pressures. JAMA 1983;249:2792-8.

173. Kikuya M, Ohkubo T, Asayama K, et al. Ambulatory blood pressureand 10-year risk of cardiovascular and noncardiovascular mortality: theOhasama study. Hypertension 2005;45:240-5.

174. Hermida RC, Smolensky MH, Ayala DE, et al. 2013 Ambulatory bloodpressure monitoring recommendations for the diagnosis of adult hy-pertension, assessment of cardiovascular and other hypertension-associated risk, and attainment of therapeutic goals (summary). Jointrecommendations from the International Society for Chronobiology(ISC), American Association of Medical Chronobiology and Chro-notherapeutics (AAMCC), Spanish Society of Applied Chronobiology,Chronotherapy, and Vascular Risk (SECAC), Spanish Society ofAtherosclerosis (SEA), and Romanian Society of Internal Medicine(RSIM) [in Spanish]. Clin Investig Arterioscler 2013;25:74-82.

175. Hermida RC, Ayala DE, Smolensky MH, et al. Chronotherapeutics ofconventional blood pressure-lowering medications: simple, low-costmeans of improving management and treatment outcomes ofhypertensive-related disorders. Curr Hypertens Rep 2014;16:412.

176. Hermida RC, Ayala DE, Smolensky MH, et al. Chronotherapy im-proves blood pressure control and reduces vascular risk in CKD. NatRev Nephrol 2013;9:358-68.

177. Hermida RC, Ayala DE, Fernandez JR, et al. Administration-timedifferences in effects of hypertension medications on ambulatory bloodpressure regulation. Chronobiol Int 2013;30:280-314.

178. Smolensky MH, Hermida RC, Ayala DE, Tiseo R, Portaluppi F.Administration-time-dependent effects of blood pressure-loweringmedications: basis for the chronotherapy of hypertension. Blood PressMonit 2010;15:173-80.

Page 13: Consequences of Circadian and Sleep Disturbances for the

872 Canadian Journal of CardiologyVolume 31 2015

179. Hermida RC, Ayala DE. Chronotherapy with the angiotensin-converting enzyme inhibitor ramipril in essential hypertension:improved blood pressure control with bedtime dosing. Hypertension2009;54:40-6.

180. Hermida RC, Ayala DE, Fernandez JR, Calvo C. Comparison of theefficacy of morning versus evening administration of telmisartan inessential hypertension. Hypertension 2007;50:715-22.

181. Hermida RC, Ayala DE, Fontao MJ, Mojon A, Fernandez JR. Chro-notherapy with valsartan/amlodipine fixed combination: improvedblood pressure control of essential hypertension with bedtime dosing.Chronobiol Int 2010;27:1287-303.

182. Hermida RC, Ayala DE, Mojon A, Fernandez JR. Influence of circadiantime of hypertension treatment on cardiovascular risk: results of theMAPEC study. Chronobiol Int 2010;27:1629-51.

183. Ayas NT, Hirsch AA, Laher I, et al. New frontiers in obstructive sleepapnoea. Clin Sci (Lond) 2014;127:209-16.

184. Kasai T, Bradley TD. Obstructive sleep apnea and heart failure: path-ophysiologic and therapeutic implications. J Am Coll Cardiol 2011;57:119-27.

185. Floras JS. Sleep apnea and cardiovascular risk. J Cardiol 2014;63:3-8.

186. Somers VK, White DP, Amin R, et al. Sleep apnea and cardiovasculardisease: an American Heart Association/American College of CardiologyFoundation Scientific Statement from the American Heart AssociationCouncil for High Blood Pressure Research Professional EducationCommittee, Council on Clinical Cardiology, Stroke Council, andCouncil on Cardiovascular Nursing. J Am Coll Cardiol 2008;52:686-717.

187. Bradley TD, Floras JS. Sleep apnea and heart failure: part I: obstructivesleep apnea. Circulation 2003;107:1671-8.

188. Bradley TD, Floras JS. Obstructive sleep apnoea and its cardiovascularconsequences. Lancet 2009;373:82-93.

189. Usui K, Bradley TD, Spaak J, et al. Inhibition of awake sympatheticnerve activity of heart failure patients with obstructive sleep apnea bynocturnal continuous positive airway pressure. J Am Coll Cardiol2005;45:2008-11.

190. Somers VK, Dyken ME, Clary MP, Abboud FM. Sympathetic neuralmechanisms in obstructive sleep apnea. J Clin Invest 1995;96:1897-904.

191. Kaneko Y, Floras JS, Usui K, et al. Cardiovascular effects of continuouspositive airway pressure in patients with heart failure and obstructivesleep apnea. N Engl J Med 2003;348:1233-41.

192. Arias MA, Garcia-Rio F, Alonso-Fernandez A, et al. Obstructive sleepapnea syndrome affects left ventricular diastolic function: effects of nasalcontinuous positive airway pressure in men. Circulation 2005;112:375-83.

193. Tkacova R, Rankin F, Fitzgerald FS, Floras JS, Bradley TD. Effects ofcontinuous positive airway pressure on obstructive sleep apnea and leftventricular afterload in patients with heart failure. Circulation 1998;98:2269-75.

194. Pepperell JC, Ramdassingh-Dow S, Crosthwaite N, et al. Ambulatoryblood pressure after therapeutic and subtherapeutic nasal continuouspositive airway pressure for obstructive sleep apnoea: a randomisedparallel trial. Lancet 2002;359:204-10.

195. Burioka N, Koyanagi S, Endo M, et al. Clock gene dysfunction inpatients with obstructive sleep apnoea syndrome. Eur Respir J 2008;32:105-12.

196. Chan CT, Floras JS, Miller JA, Richardson RM, Pierratos A. Regressionof left ventricular hypertrophy after conversion to nocturnal hemodi-alysis. Kidney Int 2002;61:2235-9.

197. Culleton BF, Walsh M, Klarenbach SW, et al. Effect of frequentnocturnal hemodialysis vs conventional hemodialysis on left ventricularmass and quality of life: a randomized controlled trial. JAMA 2007;298:1291-9.

198. Chan CT, Arab S, Carasso S, et al. Impact of frequent nocturnal he-modialysis on myocardial mechanics and cardiomyocyte gene expres-sion. Circ Cardiovasc Imaging 2012;5:474-80.

199. Bonten TN, Saris A, van Oostrom MJ, et al. Effect of aspirin intake atbedtime versus on awakening on circadian rhythm of platelet reactivity.A randomised cross-over trial. Thromb Haemost 2014;112:1209-18.

200. Heart and Stroke Foundation of Canada. Statistics. Stroke. Available at:http://www.heartandstroke.com/site/c.ikIQLcMWJtE/b.3483991/k.34A8/Statistics.htm#stroke. Accessed August 20, 2014.