Next Article in Journal
4,6′-Anhydrooxysporidinone from Fusarium lateritium SSF2 Induces Autophagic and Apoptosis Cell Death in MCF-7 Breast Cancer Cells
Next Article in Special Issue
Clocks, Rhythms, Sex, and Hearts: How Disrupted Circadian Rhythms, Time-of-Day, and Sex Influence Cardiovascular Health
Previous Article in Journal
Inhibition of Glycine Re-Uptake: A Potential Approach for Treating Pain by Augmenting Glycine-Mediated Spinal Neurotransmission and Blunting Central Nociceptive Signaling
Previous Article in Special Issue
Assessment of Selected Clock Proteins (CLOCK and CRY1) and Their Relationship with Biochemical, Anthropometric, and Lifestyle Parameters in Hypertensive Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Circadian Blood Pressure Rhythm in Cardiovascular and Renal Health and Disease

1
Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
2
School of Bioengineering, Dalian University of Technology, Dalian 116024, China
*
Author to whom correspondence should be addressed.
Biomolecules 2021, 11(6), 868; https://doi.org/10.3390/biom11060868
Submission received: 13 May 2021 / Revised: 5 June 2021 / Accepted: 8 June 2021 / Published: 11 June 2021
(This article belongs to the Special Issue Therapeutic Aspects of Circadian Rhythms)

Abstract

:
Blood pressure (BP) follows a circadian rhythm, it increases on waking in the morning and decreases during sleeping at night. Disruption of the circadian BP rhythm has been reported to be associated with worsened cardiovascular and renal outcomes, however the underlying molecular mechanisms are still not clear. In this review, we briefly summarized the current understanding of the circadian BP regulation and provided therapeutic overview of the relationship between circadian BP rhythm and cardiovascular and renal health and disease.

1. Introduction of Circadian BP Rhythm

Circadian rhythm refers to an endogenous biorhythm with a 24 h cycle driven by an intrinsic and periodic molecular clock that adjusts behavior and physiological activities to external environmental changes [1,2]. In mammals, the master pacemaker located in the suprachiasmatic nucleus (SCN) of the hypothalamus elaborately controls the peripheral clocks in other tissues to ensure the synchronization of all existing clocks [3,4]. As is shown in Figure 1, the molecular circadian clock in mammals is driven by interactional transcription-translation auto-regulatory feedback loop with a subset of core clock genes including Bmal1 (brain and muscle aryl-hydrocarbon receptor nuclear translocator-like 1), Clock (circadian locomotor output cycles kaput), Per1/2/3 (period1/2/3) and Cry1/2 (cryptochrome1/2). BMAL1 and CLOCK proteins collectively form a heterodimer to bind on E-box elements in promoter regions of clock-controlled genes (CCGs) including several important genes related to cardiovascular and renal function [5,6]. When the accumulation of PER and CRY proteins is fully superfluous in the cytoplasm, both of them translocate to the nucleus where shut down the transcription of heterodimeric activator [7]. In addition, nuclear receptors ROR (RAR-related orphan receptor) activates and REV-ERB (nuclear receptor subfamily 1, group D) suppresses RORE (ROR element) to directly control Bmal1 and Clock transcription [8]. Moreover, another nuclear receptor PPARγ integrated in this loop is also a target gene of BMAL1 by directly binding to PPRE (PPAR response element) in its promoter [9]. Particularly, all of dynamic but coordinated PPAR isoforms displayed diurnal expression at different time point in mouse tissues [10]. It is estimated that almost 10–15% of the gene transcripts in the heart and kidney are under circadian transcriptional control [11].
Emerging evidence indicates that most cardiovascular and renal functional fluctuations are evolutionarily controlled by circadian clocks. BP, which is crucial to cardiovascular and renal health, displays obvious circadian rhythm with nocturnal dip and morning surge in both human and rodents [12]. The circadian pattern of BP can be affected by many factors.
For instance, melatonin, secreted from the pineal gland at night, may significantly reduce nocturnal BP and improve sleep in patients with essential hypertension [13]. Rhythmicity of atrial natriuretic peptide (ANP) appeared to be in antiphase with BP rhythm, which has been proven to serve as an important regulatory factor of the 24 h BP pattern and affect the cardiovascular system [14]. Circadian rhythms of plasma renin activity (PRA), angiotensin converting enzyme (ACE) activity, concentration of AngII, aldosterone and thyroid hormone contribute to the maintaining 24 h BP rhythm as well [15,16,17,18,19]. In addition to hormones, blood vessel components, such as smooth muscle cells and endothelial cells, and many vasoactive substances can change the diurnal variations of BP. Typically, nitric oxide (NO) secreted from endothelial cells modulates vascular tone and thus BP. NO production is increased in the morning, followed by a morning surge in BP. Yet, whether the diurnal variation of NO is directly related to BP rhythmicity is in debate. Disruption of the diurnal oscillation of NO production has been closely connected with BP alterations in cardiovascular disease [20]. Furthermore, endothelial NO synthase (eNOS) is one of the three NO synthase enzymes, which produce NO in blood vessels and regulate vascular function. In animal studies, the phosphorylated-eNOS (p-eNOS) in the blood vessels of young mice exhibited a circadian rhythm. The core clock genes can also regulate the coupling of eNOS and contribute to the maintenance of rhythmicity of endothelial function and BP [21]. Similarly, although 3.5-day rhythm and 8-hourly change have been reported in the human circulation [22], the secretion of endothelin 1 (ET-1) in plasma and excretion in urine has been also pronouncedly confirmed to be circadian rhythmic and closely related with BP homeostasis [23,24,25]. ET-1 receptor antagonists had been proved to be efficient for the treatment of essential hypertension, however, significant fluid retention and edema side effects were also reported [26]. Besides the above, there are many other endogenous factors contribute to the 24 h BP oscillations through humoral, endocrine, neural or other coordinated regulation signals. Furthermore, some diseases can alter the 24 h BP rhythm. For instance, patients with obstructive sleep apnea (OSA) are more likely to have non-dipper BP, which will increase the incidence of cardiovascular events [27].
For exogenous aspect, growing studies have revealed that the sleep patterns may have significant effects on BP day-night profile [28]. For instance, people with sleep disturbance or circadian misalignment by shift work and social jet lag often suffer from hypertension [29], BP phase delay [30], abnormal rhythm of melatonin secretion [31] or increased high-sensitivity C-reactive protein [32]. Furthermore, although our own studies have illustrated that the diurnal rhythms of BP are subject to more direct control by the clock than behavior, at least under basal conditions [33,34]. The most recent study, however, found that the timing of food intake can entrain the diurnal BP rhythm independent to Bmal1 and the light-dark cycles [35]. In addition, the rhythm of BP can be affected by temperature, noise and many other environmental factors [36,37,38].

2. Canonical Clock Genes and BP Regulation

As mentioned above, the circadian rhythm of BP is driven by a complicated molecular clock network. Growing evidence from rodents has found that the circadian rhythms of BP can be distinctly altered by genetic manipulation of the core clock genes (Table 1).

2.1. Bmal1

Several studies indicated that mice of Bmal1 deletion exhibited altered BP and (or) impaired cardiovascular and renal functions [21,39,48,51,52,54,55,56]. Loss of diurnal variation in BP accompanied by hypotension phenotype has been confirmed in both prenatal and postnatal Bmal1 global knockout mice [33,39]. On the contrary, although Bmal1 knockout rats resulted in an overall lowering of arterial pressure, the BP rhythms remained intact [40]. Differences between species may be a possible explanation. Specific depletion of Bmal1 in mice from smooth muscle cells (SM22α-Cre), but not from cardiomyocytes, significantly suppressed the amplitude and shifted the acrophase of BP oscillation [48]. Mechanistically, BMAL1 regulates the time-of-day variations of Rho-kinase 2 (ROCK2) activity by directly binding on its promoter, and Bmal1 deletion greatly diminished ROCK2 activation, myosin phosphorylation, and then the diurnal variations in response to vasoconstriction [48]. Chang and colleagues showed that loss of Bmal1 in perivascular adipose tissue reduced BP in mice during the resting phase [49]. Moreover, mice with depression of Bmal1 in endothelial cells (TEK-Cre) had significantly lowered mean arterial pressure as well, however the molecular mechanism is still not clear [50]. In terms of renal Bmal1 in circadian BP regulation, Firsov and colleagues have confirmed a mild decrease in systolic BP, but not diastolic BP, with intact circadian rhythm in the mice lacking Bmal1 in the entire nephron (Pax8-Cre) [51]. While in another mouse model where Bmal1 was depleted in the renin-secreting cells (Ren1-Cre), reduction in the amplitude of BP was also observed, which is accompanied with many other renal abnormalities including the changes in the circadian rhythm of urinary sodium excretion and increased glomerular filtration rate, and reduced plasma aldosterone levels [52]. In addition, Pollock and colleagues have shown that loss of Bmal1 in the collecting duct (AQP2-Cre) significantly lowers BP in male, but not female mice compared to control mice [53]. Gumz and colleagues showed mice lacking Bmal1 in the thick ascending limb, distal convoluted tubule, and collecting duct cells (Ksp-Cre) displayed an obviously lower basal systolic BP compared to control mice [54]. Taken together, Bmal1, especially expression in cardiovascular and renal system, plays critical role in maintaining BP homeostasis and rhythm.

2.2. Per

Most recently, a series of studies from rodents have elegantly clarified the important role of Per1 in BP regulation and electrolyte handling. Radiotelemetry recordings and Elisa measurement showed that mice lacking Per1 exhibited significantly reduced BP and elevated renal ET-1 levels in male 129/sv mice [43]. However, altered ET-1 levels were not observed in female Per1 knockout mice with the C57BL/6 background [57]. High salt diet plus mineralocorticoid deoxycorticosterone pivalate (HS/DOCP) injection significantly resulted in non-dipping hypertension and renal sodium handling defect (<10% difference between active and inactive phase) in male Per1 knockout mice [44,58]. Interestingly, the female Per1 knockout mice appeared to be protective against non-dipping hypertension in response to HS/DOCP treatment in sex-dependent manner to some extent [57]. Moreover, in contrast to C57BL/6 background, male Per1 KO mice on a 129/sv background in combination with HS/DOCP challenge are extraordinarily protective from hypertension [59], implying that sexual hormones might be fundamental to Per1 medicated circadian BP control. The complexity of Per1 in circadian BP control may need further investigation. In accordance with altered BP pattern, the clock gene Per1 might regulate the transcription of epithelial sodium channel (ENaC), sodium-glucose linked transporter-1 (SGLT-1), sodium-hydrogen exchanger-3 (NHE3), endothelin-1 (ET-1), which are responsible for sodium transport in the kidney [43,60,61]. As for another isoform of gene Per, Per2 mutant mice showed a mild cardiovascular phenotype with an elevated 24 h heart rate as well as decreased 24 h diastolic BP during the active phase [46]. However, the systolic BP and mean arterial BP displayed no significant difference between the mutant and control mice [46]. In addition, Per2 mutant mice resulted in mild attenuation of 24 h BP rhythm, heart rate, and locomotor activity with much shorter circadian periods than wild-type mice under constant dark condition [46]. Collectively, the clock component Per acts as a crucial mediator in circadian system to be involved in BP regulation.

2.3. Clock and Cry

It has been reported that the Clock mutant mice on the Jcl/ICR background displayed dampened diurnal rhythm of BP and heart rate [42]. While in Clock knockout mice, the arterial BP was normally rhythmic and significantly lower with about 10 mmHg decline compared to control mice, accompanied by mild diabetes insipidus, impaired sodium excretion rhythm [41]. With regard to another core clock component Cry, Doi et al. have unveiled that simultaneously lacking Cry1 and Cry2 in mice would lead to salt-sensitive hypertension due to abnormally chronic over-production of the mineralocorticoid aldosterone by the disordered adrenal gland [47]. DNA microarray further revealed that overexpression of a specific enzyme Hsd3b6 may contribute to the high mineralocorticoid aldosterone synthesis in Cry-null mice, implying that clock-dependent sodium excretion may play an important role in the BP circadian homeostasis [47]. However, the causal connection between altered sodium excretion and BP regulation needs to be further explored.
It is worth mentioning that in addition to the core clock gene manipulation models, the transgenic hypertensive TGR(mREN2)27 rat, which harbors an additional mouse renin gene and exhibits an inverse circadian BP profile, has also been commonly used for circadian BP rhythm studies [62,63,64].

3. Disruption of Circadian BP Rhythm and Cardiovascular Disease

BP rhythm is one of the most common circadian rhythms in the cardiovascular system [65]. Disruption of the BP rhythm has been considered to be a major contribution to many adverse cardiovascular events. It has been found that every 5% drop in the decline in nocturnal BP may increase the risk of cardiovascular death by about 20% [66].
Myocardial infarction (MI) is an acute heart disease, which can be complicated by arrhythmia, shock or heart failure, and is life-threatening. There is an inseparable connection between the rhythm of BP and MI. Abnormal BP circadian patterns or chronic hypertension may lead to changes in cardiac hemodynamics, and increase the incidence of MI [67]. Several Clinical studies have proved that the incidence of MI is rhythmic and significantly increased incidence rate was usually observed between 06:00 am and 12:00 pm which is somehow consistent to the BP morning surge. As for systolic BP, lower (<100 mm Hg) in post-MI period increase the risk of cardiovascular events, while patients with elevated systolic BP (>140 mm Hg) have a higher risk of stroke and combined cardiovascular events [68]. Compared with resting systolic BP, systolic BP after exercise can provide higher predictive value for acute MI [69]. In terms of diastolic BP, acute myocardial infarction (AMI) patients with lower diastolic BP (<70 mmHg) have increased postoperative risks, including all-cause death, cardiovascular death and cardiovascular hospitalization. Moreover, wide pulse pressure was also considered as a predictor of MI [70]. Furthermore, several core clock genes are considered as important components in regulating the rhythmic pathogenesis of MI. There is an association between MI and Clock and Arntl (also known as Bmal1) gene polymorphisms [71], and the genetic variants of Arntl, Clock and Per2 genes are proved to be related to circadian phenotype (i.e., chronotype or daytime sleepiness) in patients with MI [72].
Under the condition of heart failure with preserved ejection fraction, the increased variability of systolic and diastolic BP is related to the increase of adverse events [73]. Concurrently, abnormal systolic BP and diastolic BP can cause the variation of heart indicators. As the systolic BP increases, the left ventricular wall thickness and left ventricular mass index also increase with the higher diastolic BP; while the functional index of left ventricular diastolic function is inversely proportional to the increase of diastolic BP, but irrelevant with systolic BP [74]. Overall, there is a U-shaped relationship between systolic BP and heart failure, that is, lower or higher systolic BP will lead to higher risk of death and heart failure hospitalization [75]. The same phenomenon exists in diastolic BP [76]. Therefore, when diagnosing MI and related cardiovascular events, it is necessary to comprehensively consider the phenotype and rhythms of BP.
Given the significant correlation between acute cardiovascular events and BP rhythms, it is necessary to improve the therapeutic strategies according to the characteristics of circadian rhythm changes. In addition to optimizing the drug administration time, it has been reported that the time of cardiovascular surgery may have influences on the tissue repair and outcome of cardiovascular diseases as well. For example, although the most recent studies demonstrated that the time-of-day variation of aortic valve replacement surgery had no significant impact on clinical outcomes and had no clinically relevant biorhythm for myocardial ischemia-reperfusion tolerance [77,78,79]. Montaigne et al. did have reported that the incidence of major adverse cardiac events in the afternoon surgery group was significantly lower than that in the morning group [80]. More interestingly, in patients undergoing non-cardiac surgery, Lavallaz et al. found that the incidence of AMI during follow-up is instead increased in the afternoon surgeries [81]. Nevertheless, a better understanding of the relationship between circadian clock and cardiovascular disease may help to develop more targeted and personalized treatment strategies for patients.

4. Disruption of Circadian BP Rhythm and Renal Damage

Chronic kidney disease has long been closely linked with sleep disturbance and hypertension and thus gradually become a worldwide health problem with a great economic burden [82,83]. In humans, accumulating clinical evidence indicated that blunted BP variation from 24 h BP recordings was associated with increased risk of end-organ damage and progressive loss of renal function [84,85]. Predictably, the consistency of circadian BP variation was ruined in chronic kidney disease stages 3–5 [86]. In addition, among patients with chronic kidney disease, circadian BP profile is closely related to the level of physical activity and the severity of target organ damage [87]. Moreover, clinical trials indicated that circadian BP profile is associated with mortality among elderly veterans with chronic kidney disease [88]. Meanwhile, the nocturnal dip in BP is nearly diminished in patients with glomerulopathy, resulting in enhanced urinary excretion rate of sodium and protein during the inactive phase [89].
To date, an explicit mechanism has not been expatiated between chronic kidney disease and disruption of circadian BP rhythm. In rodent models, studies have found that after treatment with adenine for 2 to 4 weeks to induce chronic kidney disease in mice or Sprague–Dawley rats, the animals will display significantly increased arterial BP and disrupted day-night variation [90]. In addition, the SCN of PER2::LUC mice with adenine-induced chronic kidney disease displayed dampened amplitude rhythms in their central circadian clock as measured by bioluminescence. Meanwhile, the kidney explants from chronic kidney disease mice expressed altered PER2::LUC rhythms with significantly longer period compared to control mice [91]. On the other hand, upon adenine treatment, Clock mutant mice showed significantly higher BP and worse renal function than control mice, implying the circadian BP disruption enhances the susceptibility to chronic kidney disease [91]. However, our own recent study found that postnatal Bmal1 deletion in mice protected against renal fibrosis via suppressing Gli2 transcription, which provides a prominent role of the core clock gene Bmal1 in tubulointerstitial fibrosis [92]. Nevertheless, more efforts need to be made from different point of view to further clarify the relationship between circadian BP disruption and chronic kidney diseases.

5. Chronotherapy of Anti-Hypertensive Medications

Altered day-night oscillation in BP has been associated with high morbidity and mortality of cardiovascular disease and increased progression of kidney damage. Of note, non-dipping hypertension is frequently observed in clinical outcomes of chronic kidney disease and associated with increased risk of cardiovascular event [93]. While an excessive circadian variation in BP is also associated with an increased risk of nephropathy [94,95]. Thus, the application of anti-hypertensive drugs, including angiotensin-converting enzyme inhibitors (ACEI), angiotensin II receptor blockers (ARB) and others, becomes fundamental to the prevention and treatment of many cardiovascular and renal diseases. The rational and effective administration time of these anti-hypertension medications has also been explored and reported in many clinical trials and animal studies.

5.1. ACEI

Clinical studies demonstrated that bedtime administration of the long-acting lipophilic ACEI trandolapril was a safer and more effective mean of morning BP control in hypertensive patients compared to morning-dosed group [96]. The combination medication of captopril and hydrochlorothiazide was mildly more effective in nighttime BP reduction when ingested in the evening in 13 male patients with moderate hypertension for 3 weeks [97]. In addition, bedtime spirapril administration was more efficient than morning administration in reducing asleep BP, which might be associated with the nocturnal RAAS activation [98]. Additionally, evening treatment of enalapril would further decrease nighttime BP followed by a slow increase during the day in a randomized crossover design [99]. Furthermore, lisinopril administration at 10.00 pm. has been shown to be much more useful [100]. With regard to quinapril, the evening administration resulted in similarly more preferable antihypertensive effect, whereas the morning schedule represented a smaller reduction in nighttime BP [101]. However, there are also examples of the opposite, in a single-blinded crossover study of 10 hypertensive subjects, which have been reported that morning administration of benazepril had a more sustained anti-hypertensive effect than evening administration [102]. Moreover, although the hemodynamics seemed better after the evening intake of the ramipril medication, the hypotensive effect of ramipril and imidapril appeared approximately the similar between morning administration and evening administration in a crossover study involving patients with hypertension [103,104].

5.2. ARB

ARBs are becoming more popular for the treatment of hypertension and highly effective and well tolerated. Valsartan administration at bedtime, as opposed to upon awakening, resulted in a highly significant average increase by 6% in the day-night BP difference toward a more dipper pattern, which corresponded to a 73% relative reduction in the number of patients with non-dipping hypertension [105,106]. Furthermore, it has been found amlodipine and valsartan combination therapy should be preferably administrated at bed time for the majority of controlled patients with potential additional cardiovascular event occurrence reduction [107]. However, in subjects with requirement to combined medication to achieve proper BP-lowering effect, co-administration of amlodipine and valsartan efficiently reduced BP for the entire 24 h independent of dosing time compared to any one of the other assessed therapies [107]. Studies also demonstrated that telmisartan administered at bedtime improved the sleep time-relative BP decline and nocturnal BP regulation compared to dosing upon awakening [108]. Olmesartan, another angiotensin II type 1 receptor blocker, was also shown to retain night-time BP dip possibly by enhancing day-time sodium excretion to relieve cardio-renal load through normalization of circadian BP rhythm [109,110,111]. However, although evidence has shown administration of olmesartan at bedtime was significantly more efficient in reducing the nocturnal BP than morning administration [112], controversial observations have also been reported in some trials [113].

5.3. Other Anti-Hypertension Medications

Several prospective trials have been reported to elucidate the protective effect on low-dose aspirin in patients suffering from hypertension only administrated at bedtime, beyond the secondary prevention of cardiovascular disease [114,115,116,117,118]. Likewise, we have recapitulated the paralleled time-dependent hypotensive effect on hypertensive mice treated with low-dose aspirin [119], which could provide a beneficial approach to solve the BP control of hypertensive subjects throughout the day. Additionally, nifedipine is also marked as an anti-hypertensive agent. The BP reduction after nifedipine treatment was significantly larger with bedtime administration than morning ingestion, as well as efficiently reduced the incidence of edema and the total number of adverse effects compared to ingestion of nifedipine on awakening [120,121,122].
Taken together, although growing evidence demonstrated that bedtime administration of the current anti-hypertension medications would be more efficiently in reducing BP, however there are also many inconsistencies. Indeed, in the most recent Hygia Chronotherapy Trial, which examined the effects of bedtime vs. upon-waking BP-lowering treatment on cardiovascular risk in a large cohort with 19,804 participating patients, although authors claimed that routine bedtime ingestion of more than one BP-lowering medications may result in improved BP control and diminished occurrence of adverse cardiovascular events [123], this study only included patients from one population, the reduction of BP was relatively small, and it seemed no properly randomized controlled trial were performed in this big trial [124]. In addition, in a trial with patients of hypertension-related chronic kidney diseases, Rahman et al. also failed to observe any difference between the bedtime and morning antihypertensive treatment on BP control [125]. These inconsistencies may reflect the differences in cohort or drug selection or the variations in BP monitoring methodologies. However, attention may have to be brought to the limitations of recommending a single optimal time for an entire population, without concern for the chronodiagnosis and effects of the anti-hypertensive medications on the circadian amplitude and phase of BP. Some, but not all anti-hypertensive medications affect the circadian amplitude of BP [126]. Nevertheless, further in-depth exploration is required for the chronotherapy of anti-hypertensive medications. Merits of personalized chronotherapy that account for the chronodiagnosis have been documented [127].

6. Conclusions and Perspectives

Growing evidence indicates that the maintenance of BP circadian rhythm is highly associated with the cardiovascular and renal homeostasis, many anti-hypertension medications displayed clear circadian time-dependent efficiency and chronotherapeutic evaluation to the management of hypertension and the related cardiovascular and renal diseases had be taken into consideration for many studies. However, further research in this field still has to lay emphasis on accurate pharmacodynamics and pharmacokinetics of time-dependent administration by using novel compounds that modulate circadian timing system to superiorly prevent the onset and progression of hypertension and the related cardiovascular and chronic kidney diseases, which will provide a valuable and cost-benefit approach for BP control and potential merit of chronotherapy, as well as added systematic protection.
Moreover, the molecular link between the circadian BP regulation and cardiovascular and renal functions is still less known. RNA-seq analysis revealed that heart and kidney are two of the top five candidates along with the liver, lung and brown fat following obvious circadian regulation. However, the complexity of the cellular components and molecular network in the heart and kidney largely renders the mechanism exploration. Further study by tissue or cell specific depletion of the circadian genes in the heart and kidney may provide a better understanding of the relationship between BP rhythm and cardiovascular and renal function and pave a way for preventing the development of cardiovascular and renal diseases.

Author Contributions

All authors have participated in the writing, review and editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Key R&D Program of China (grant number: #2019YFA0802400) and National Natural Science Foundation of China (grant number: 32071157 and 31871190).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Roenneberg, T.; Merrow, M. Circadian clocks—The fall and rise of physiology. Nat. Rev. Mol. Cell Biol. 2005, 6, 965–971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Harmer, S.L.; Panda, S.; Kay, S.A. Molecular bases of circadian rhythms. Annu. Rev. Cell Dev. Biol. 2001, 17, 215–253. [Google Scholar] [CrossRef] [Green Version]
  3. Lowrey, P.L.; Takahashi, J.S. Genetics of circadian rhythms in Mammalian model organisms. Adv. Genet. 2011, 74, 175–230. [Google Scholar] [CrossRef] [Green Version]
  4. Dibner, C.; Schibler, U.; Albrecht, U. The mammalian circadian timing system: Organization and coordination of central and peripheral clocks. Annu. Rev. Physiol. 2010, 72, 517–549. [Google Scholar] [CrossRef] [Green Version]
  5. Zhang, D.; Pollock, D.M. Diurnal regulation of renal electrolyte excretion: The role of paracrine factors. Annu. Rev. Physiol. 2019. [Google Scholar] [CrossRef] [Green Version]
  6. Lutshumba, J.; Liu, S.; Zhong, Y.; Hou, T.; Daugherty, A.; Lu, H.; Guo, Z.; Gong, M.C. Deletion of BMAL1 in smooth muscle cells protects mice from abdominal aortic aneurysms. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 1063–1075. [Google Scholar] [CrossRef] [Green Version]
  7. Duong, H.A.; Robles, M.S.; Knutti, D.; Weitz, C.J. A molecular mechanism for circadian clock negative feedback. Science 2011, 332, 1436–1439. [Google Scholar] [CrossRef] [Green Version]
  8. Guillaumond, F.; Dardente, H.; Giguere, V.; Cermakian, N. Differential control of Bmal1 circadian transcription by REV-ERB and ROR nuclear receptors. J. Biol. Rhythm. 2005, 20, 391–403. [Google Scholar] [CrossRef]
  9. Wang, N.; Yang, G.; Jia, Z.; Zhang, H.; Aoyagi, T.; Soodvilai, S.; Symons, J.D.; Schnermann, J.B.; Gonzalez, F.J.; Litwin, S.E.; et al. Vascular PPARgamma controls circadian variation in blood pressure and heart rate through Bmal1. Cell Metab. 2008, 8, 482–491. [Google Scholar] [CrossRef] [Green Version]
  10. Yang, X.; Downes, M.; Yu, R.T.; Bookout, A.L.; He, W.; Straume, M.; Mangelsdorf, D.J.; Evans, R.M. Nuclear receptor expression links the circadian clock to metabolism. Cell 2006, 126, 801–810. [Google Scholar] [CrossRef] [Green Version]
  11. Zhang, R.; Lahens, N.F.; Ballance, H.I.; Hughes, M.E.; Hogenesch, J.B. A circadian gene expression atlas in mammals: Implications for biology and medicine. Proc. Natl. Acad. Sci. USA 2014, 111, 16219–16224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Smolensky, M.H.; Hermida, R.C.; Portaluppi, F. Circadian mechanisms of 24-hour blood pressure regulation and patterning. Sleep Med. Rev. 2017, 33, 4–16. [Google Scholar] [CrossRef] [PubMed]
  13. Scheer, F.A.; Van Montfrans, G.A.; van Someren, E.J.; Mairuhu, G.; Buijs, R.M. Daily nighttime melatonin reduces blood pressure in male patients with essential hypertension. Hypertension 2004, 43, 192–197. [Google Scholar] [CrossRef] [Green Version]
  14. Portaluppi, F.; Montanari, L.; Bagni, B.; degli Uberti, E.; Trasforini, G.; Margutti, A. Circadian rhythms of atrial natriuretic peptide, blood pressure and heart rate in normal subjects. Cardiology 1989, 76, 428–432. [Google Scholar] [CrossRef] [PubMed]
  15. Gordon, R.D.; Wolfe, L.K.; Island, D.P.; Liddle, G.W. A diurnal rhythm in plasma renin activity in man. J. Clin. Investig. 1966, 45, 1587–1592. [Google Scholar] [CrossRef] [PubMed]
  16. Sica, D.A. What are the influences of salt, potassium, the sympathetic nervous system, and the renin-angiotensin system on the circadian variation in blood pressure? Blood Press. Monit. 1999, 4 (Suppl. 2), S9–S16. [Google Scholar]
  17. Veglio, F.; Pietrandrea, R.; Ossola, M.; Vignani, A.; Angeli, A. Circadian rhythm of the angiotensin converting enzyme (ACE) activity in serum of healthy adult subjects. Chronobiologia 1987, 14, 21–25. [Google Scholar] [PubMed]
  18. Li, H.; Sun, N.L.; Wang, J.; Liu, A.J.; Su, D.F. Circadian expression of clock genes and angiotensin II type 1 receptors in suprachiasmatic nuclei of sinoaortic-denervated rats. Acta Pharmacol. Sin. 2007, 28, 484–492. [Google Scholar] [CrossRef]
  19. Liebau, H.; Manitius, J. Diurnal and daily variations of PRA, plasma catecholamines and blood pressure in normotensive and hypertensive man. Contrib. Nephrol. 1982, 30, 57–63. [Google Scholar] [CrossRef]
  20. Bode-Boger, S.M.; Boger, R.H.; Kielstein, J.T.; Loffler, M.; Schaffer, J.; Frolich, J.C. Role of endogenous nitric oxide in circadian blood pressure regulation in healthy humans and in patients with hypertension or atherosclerosis. J. Investig. Med. 2000, 48, 125–132. [Google Scholar]
  21. Anea, C.B.; Cheng, B.; Sharma, S.; Kumar, S.; Caldwell, R.W.; Yao, L.; Ali, M.I.; Merloiu, A.M.; Stepp, D.W.; Black, S.M.; et al. Increased superoxide and endothelial NO synthase uncoupling in blood vessels of Bmal1-knockout mice. Circ. Res. 2012, 111, 1157–1165. [Google Scholar] [CrossRef] [Green Version]
  22. Katinas, G.S.; Halberg, F.; Cornelissen, G.; Hawkins, D.; Bueva, M.V.; Korzhevsky, D.E.; Sapozhnikova, L.R.; Rhodus, N.; Schaffer, E. About 8- and approximately 84-h rhythms in endotheliocytes as in endothelin-1 and effect of trauma. Peptides 2001, 22, 647–659. [Google Scholar] [CrossRef]
  23. Thosar, S.S.; Berman, A.M.; Herzig, M.X.; McHill, A.W.; Bowles, N.P.; Swanson, C.M.; Clemons, N.A.; Butler, M.P.; Clemons, A.A.; Emens, J.S.; et al. Circadian rhythm of vascular function in midlife adults. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 1203–1211. [Google Scholar] [CrossRef]
  24. Hwang, Y.S.; Hsieh, T.J.; Lee, Y.J.; Tsai, J.H. Circadian rhythm of urinary endothelin-1 excretion in mild hypertensive patients. Am. J. Hypertens. 1998, 11, 1344–1351. [Google Scholar] [CrossRef] [Green Version]
  25. Soliman, R.H.; Johnston, J.G.; Gohar, E.Y.; Taylor, C.M.; Pollock, D.M. Greater natriuretic response to ENaC inhibition in male versus female Sprague-Dawley rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2020, 318, R418–R427. [Google Scholar] [CrossRef] [PubMed]
  26. Rautureau, Y.; Schiffrin, E.L. Endothelin in hypertension: An update. Curr. Opin. Nephrol. Hypertens. 2012, 21, 128–136. [Google Scholar] [CrossRef] [PubMed]
  27. Cuspidi, C.; Tadic, M.; Sala, C.; Gherbesi, E.; Grassi, G.; Mancia, G. Blood pressure non-dipping and obstructive sleep apnea syndrome: A meta-analysis. J. Clin. Med. 2019, 8, 1367. [Google Scholar] [CrossRef] [Green Version]
  28. Kario, K.; Schwartz, J.E.; Pickering, T.G. Ambulatory physical activity as a determinant of diurnal blood pressure variation. Hypertension 1999, 34, 685–691. [Google Scholar] [CrossRef] [Green Version]
  29. Riegel, B.; Daus, M.; Lozano, A.J.; Malone, S.K.; Patterson, F.; Hanlon, A.L. Shift workers have higher blood pressure medicine use, but only when they are short sleepers: A longitudinal UK biobank study. J. Am. Heart Assoc. 2019, 8, e013269. [Google Scholar] [CrossRef] [PubMed]
  30. Motohashi, Y.; Higuchi, S.; Maeda, A.; Liu, Y.; Yuasa, T.; Motohashi, K.; Nakamura, K. Alteration of circadian time structure of blood pressure caused by night shift schedule. Occup. Med. 1998, 48, 523–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Gumenyuk, V.; Roth, T.; Drake, C.L. Circadian phase, sleepiness, and light exposure assessment in night workers with and without shift work disorder. Chronobiol. Int. 2012, 29, 928–936. [Google Scholar] [CrossRef]
  32. Morris, C.J.; Purvis, T.E.; Mistretta, J.; Hu, K.; Scheer, F. Circadian misalignment increases c-reactive protein and blood pressure in chronic shift workers. J. Biol. Rhythm. 2017, 32, 154–164. [Google Scholar] [CrossRef] [Green Version]
  33. Yang, G.; Chen, L.; Grant, G.R.; Paschos, G.; Song, W.L.; Musiek, E.S.; Lee, V.; McLoughlin, S.C.; Grosser, T.; Cotsarelis, G.; et al. Timing of expression of the core clock gene Bmal1 influences its effects on aging and survival. Sci. Transl. Med. 2016, 8, 324ra316. [Google Scholar] [CrossRef] [Green Version]
  34. Yang, G.; Jia, Z.; Aoyagi, T.; McClain, D.; Mortensen, R.M.; Yang, T. Systemic PPARgamma deletion impairs circadian rhythms of behavior and metabolism. PLoS ONE 2012, 7, e38117. [Google Scholar] [CrossRef] [Green Version]
  35. Zhang, D.; Colson, J.C.; Jin, C.; Becker, B.K.; Rhoads, M.K.; Pati, P.; Neder, T.H.; King, M.A.; Valcin, J.A.; Tao, B.; et al. Timing of food intake drives the circadian rhythm of blood pressure. Function 2021, 2, zqaa034. [Google Scholar] [CrossRef]
  36. D’Elia, L.; La Fata, E.; Giaquinto, A.; Strazzullo, P.; Galletti, F. Effect of dietary salt restriction on central blood pressure: A systematic review and meta-analysis of the intervention studies. J. Clin. Hypertens. 2020, 22, 814–825. [Google Scholar] [CrossRef] [Green Version]
  37. Wang, Q.; Li, C.; Guo, Y.; Barnett, A.G.; Tong, S.; Phung, D.; Chu, C.; Dear, K.; Wang, X.; Huang, C. Environmental ambient temperature and blood pressure in adults: A systematic review and meta-analysis. Sci. Total Environ. 2017, 575, 276–286. [Google Scholar] [CrossRef] [PubMed]
  38. Chang, T.Y.; Lai, Y.A.; Hsieh, H.H.; Lai, J.S.; Liu, C.S. Effects of environmental noise exposure on ambulatory blood pressure in young adults. Environ. Res. 2009, 109, 900–905. [Google Scholar] [CrossRef] [PubMed]
  39. Curtis, A.M.; Cheng, Y.; Kapoor, S.; Reilly, D.; Price, T.S.; Fitzgerald, G.A. Circadian variation of blood pressure and the vascular response to asynchronous stress. Proc. Natl. Acad. Sci. USA 2007, 104, 3450–3455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Johnston, J.G.; Speed, J.S.; Becker, B.K.; Kasztan, M.; Soliman, R.H.; Rhoads, M.K.; Tao, B.; Jin, C.; Geurts, A.M.; Hyndman, K.A.; et al. Diurnal control of blood pressure is uncoupled from sodium excretion. Hypertension 2020, 75, 1624–1634. [Google Scholar] [CrossRef] [PubMed]
  41. Zuber, A.M.; Centeno, G.; Pradervand, S.; Nikolaeva, S.; Maquelin, L.; Cardinaux, L.; Bonny, O.; Firsov, D. Molecular clock is involved in predictive circadian adjustment of renal function. Proc. Natl. Acad. Sci. USA 2009, 106, 16523–16528. [Google Scholar] [CrossRef] [Green Version]
  42. Sei, H.; Oishi, K.; Chikahisa, S.; Kitaoka, K.; Takeda, E.; Ishida, N. Diurnal amplitudes of arterial pressure and heart rate are dampened in clock mutant mice and adrenalectomized mice. Endocrinology 2008, 149, 3576–3580. [Google Scholar] [CrossRef] [Green Version]
  43. Stow, L.R.; Richards, J.; Cheng, K.Y.; Lynch, I.J.; Jeffers, L.A.; Greenlee, M.M.; Cain, B.D.; Wingo, C.S.; Gumz, M.L. The circadian protein period 1 contributes to blood pressure control and coordinately regulates renal sodium transport genes. Hypertension 2012, 59, 1151–1156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Solocinski, K.; Holzworth, M.; Wen, X.; Cheng, K.Y.; Lynch, I.J.; Cain, B.D.; Wingo, C.S.; Gumz, M.L. Desoxycorticosterone pivalate-salt treatment leads to non-dipping hypertension in Per1 knockout mice. Acta Physiol. 2017, 220, 72–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Pati, P.; Fulton, D.J.; Bagi, Z.; Chen, F.; Wang, Y.; Kitchens, J.; Cassis, L.A.; Stepp, D.W.; Rudic, R.D. Low-salt diet and circadian dysfunction synergize to induce angiotensin II-dependent hypertension in mice. Hypertension 2016, 67, 661–668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Vukolic, A.; Antic, V.; Van Vliet, B.N.; Yang, Z.; Albrecht, U.; Montani, J.P. Role of mutation of the circadian clock gene Per2 in cardiovascular circadian rhythms. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2010, 298, R627–R634. [Google Scholar] [CrossRef] [Green Version]
  47. Doi, M.; Takahashi, Y.; Komatsu, R.; Yamazaki, F.; Yamada, H.; Haraguchi, S.; Emoto, N.; Okuno, Y.; Tsujimoto, G.; Kanematsu, A.; et al. Salt-sensitive hypertension in circadian clock-deficient Cry-null mice involves dysregulated adrenal Hsd3b6. Nat. Med. 2010, 16, 67–74. [Google Scholar] [CrossRef]
  48. Xie, Z.; Su, W.; Liu, S.; Zhao, G.; Esser, K.; Schroder, E.A.; Lefta, M.; Stauss, H.M.; Guo, Z.; Gong, M.C. Smooth-muscle BMAL1 participates in blood pressure circadian rhythm regulation. J. Clin. Investig. 2015, 125, 324–336. [Google Scholar] [CrossRef] [Green Version]
  49. Chang, L.; Xiong, W.; Zhao, X.; Fan, Y.; Guo, Y.; Garcia-Barrio, M.; Zhang, J.; Jiang, Z.; Lin, J.D.; Chen, Y.E. Bmal1 in perivascular adipose tissue regulates resting-phase blood pressure through transcriptional regulation of angiotensinogen. Circulation 2018, 138, 67–79. [Google Scholar] [CrossRef]
  50. Westgate, E.J.; Cheng, Y.; Reilly, D.F.; Price, T.S.; Walisser, J.A.; Bradfield, C.A.; FitzGerald, G.A. Genetic components of the circadian clock regulate thrombogenesis in vivo. Circulation 2008, 117, 2087–2095. [Google Scholar] [CrossRef] [Green Version]
  51. Nikolaeva, S.; Ansermet, C.; Centeno, G.; Pradervand, S.; Bize, V.; Mordasini, D.; Henry, H.; Koesters, R.; Maillard, M.; Bonny, O.; et al. Nephron-specific deletion of circadian clock gene bmal1 alters the plasma and renal metabolome and impairs drug disposition. J. Am. Soc. Nephrol. 2016, 27, 2997–3004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Tokonami, N.; Mordasini, D.; Pradervand, S.; Centeno, G.; Jouffe, C.; Maillard, M.; Bonny, O.; Gachon, F.; Gomez, R.A.; Sequeira-Lopez, M.L.; et al. Local renal circadian clocks control fluid-electrolyte homeostasis and BP. J. Am. Soc. Nephrol. 2014, 25, 1430–1439. [Google Scholar] [CrossRef] [Green Version]
  53. Zhang, D.; Jin, C.; Obi, I.E.; Rhoads, M.K.; Soliman, R.H.; Sedaka, R.S.; Allan, J.M.; Tao, B.; Speed, J.S.; Pollock, J.S.; et al. Loss of circadian gene Bmal1 in the collecting duct lowers blood pressure in male, but not female, mice. Am. J. Physiol. Ren. Physiol. 2020, 318, F710–F719. [Google Scholar] [CrossRef] [Green Version]
  54. Crislip, G.R.; Douma, L.G.; Masten, S.H.; Cheng, K.Y.; Lynch, I.J.; Johnston, J.G.; Barral, D.; Glasford, K.B.; Holzworth, M.R.; Verlander, J.W.; et al. Differences in renal BMAL1 contribution to sodium homeostasis and blood pressure control in male and female mice. Am. J. Physiol. Ren. Physiol. 2020. [Google Scholar] [CrossRef] [PubMed]
  55. Ingle, K.A.; Kain, V.; Goel, M.; Prabhu, S.D.; Young, M.E.; Halade, G.V. Cardiomyocyte-specific Bmal1 deletion in mice triggers diastolic dysfunction, extracellular matrix response, and impaired resolution of inflammation. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H1827–H1836. [Google Scholar] [CrossRef]
  56. Lefta, M.; Campbell, K.S.; Feng, H.Z.; Jin, J.P.; Esser, K.A. Development of dilated cardiomyopathy in Bmal1-deficient mice. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, H475–H485. [Google Scholar] [CrossRef] [Green Version]
  57. Douma, L.G.; Solocinski, K.; Holzworth, M.R.; Crislip, G.R.; Masten, S.H.; Miller, A.H.; Cheng, K.Y.; Lynch, I.J.; Cain, B.D.; Wingo, C.S.; et al. Female C57BL/6J mice lacking the circadian clock protein PER1 are protected from nondipping hypertension. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2019, 316, R50–R58. [Google Scholar] [CrossRef]
  58. Douma, L.G.; Holzworth, M.R.; Solocinski, K.; Masten, S.H.; Miller, A.H.; Cheng, K.Y.; Lynch, I.J.; Cain, B.D.; Wingo, C.S.; Gumz, M.L. Renal Na-handling defect associated with PER1-dependent nondipping hypertension in male mice. Am. J. Physiol. Ren. Physiol. 2018, 314, F1138–F1144. [Google Scholar] [CrossRef]
  59. Alli, A.; Yu, L.; Holzworth, M.; Richards, J.; Cheng, K.Y.; Lynch, I.J.; Wingo, C.S.; Gumz, M.L. Direct and indirect inhibition of the circadian clock protein Per1: Effects on ENaC and blood pressure. Am. J. Physiol. Ren. Physiol. 2019, 316, F807–F813. [Google Scholar] [CrossRef]
  60. Gumz, M.L.; Stow, L.R.; Lynch, I.J.; Greenlee, M.M.; Rudin, A.; Cain, B.D.; Weaver, D.R.; Wingo, C.S. The circadian clock protein Period 1 regulates expression of the renal epithelial sodium channel in mice. J. Clin. Investig. 2009, 119, 2423–2434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Solocinski, K.; Richards, J.; All, S.; Cheng, K.Y.; Khundmiri, S.J.; Gumz, M.L. Transcriptional regulation of NHE3 and SGLT1 by the circadian clock protein Per1 in proximal tubule cells. Am. J. Physiol. Ren. Physiol. 2015, 309, F933–F942. [Google Scholar] [CrossRef] [Green Version]
  62. Lemmer, B.; Mattes, A.; Bohm, M.; Ganten, D. Circadian blood pressure variation in transgenic hypertensive rats. Hypertension 1993, 22, 97–101. [Google Scholar] [CrossRef] [Green Version]
  63. Lemmer, B.; Witte, K.; Enzminger, H.; Schiffer, S.; Hauptfleisch, S. Transgenic TGR(mREN2)27 rats as a model for disturbed circadian organization at the level of the brain, the heart, and the kidneys. Chronobiol. Int. 2003, 20, 711–738. [Google Scholar] [CrossRef]
  64. Zeman, M.; Szantoova, K.; Stebelova, K.; Mravec, B.; Herichova, I. Effect of rhythmic melatonin administration on clock gene expression in the suprachiasmatic nucleus and the heart of hypertensive TGR(mRen2)27 rats. J. Hypertens. Suppl. 2009, 27, S21–S26. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, L.; Yang, G. Recent advances in circadian rhythms in cardiovascular system. Front. Pharmacol. 2015, 6, 71. [Google Scholar] [CrossRef] [Green Version]
  66. Ohkubo, T.; Hozawa, A.; Yamaguchi, J.; Kikuya, M.; Ohmori, K.; Michimata, M.; Matsubara, M.; Hashimoto, J.; Hoshi, H.; Araki, T.; et al. Prognostic significance of the nocturnal decline in blood pressure in individuals with and without high 24-h blood pressure: The Ohasama study. J. Hypertens. 2002, 20, 2183–2189. [Google Scholar] [CrossRef]
  67. Virag, J.A.; Lust, R.M. Circadian influences on myocardial infarction. Front. Physiol. 2014, 5, 422. [Google Scholar] [CrossRef] [PubMed]
  68. Thune, J.J.; Signorovitch, J.; Kober, L.; Velazquez, E.J.; McMurray, J.J.; Califf, R.M.; Maggioni, A.P.; Rouleau, J.L.; Howlett, J.; Zelenkofske, S.; et al. Effect of antecedent hypertension and follow-up blood pressure on outcomes after high-risk myocardial infarction. Hypertension 2008, 51, 48–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Laukkanen, J.A.; Kurl, S.; Salonen, R.; Lakka, T.A.; Rauramaa, R.; Salonen, J.T. Systolic blood pressure during recovery from exercise and the risk of acute myocardial infarction in middle-aged men. Hypertension 2004, 44, 820–825. [Google Scholar] [CrossRef] [Green Version]
  70. Safar, M. Relation of pulse pressure and blood pressure reduction to the incidence of myocardial infarction. Hypertension 1994, 24, 368. [Google Scholar] [CrossRef] [Green Version]
  71. Skrlec, I.; Milic, J.; Steiner, R. The impact of the circadian genes CLOCK and ARNTL on myocardial infarction. J. Clin. Med. 2020, 9, 484. [Google Scholar] [CrossRef] [Green Version]
  72. Skrlec, I.; Milic, J.; Heffer, M.; Wagner, J.; Peterlin, B. Circadian clock genes and circadian phenotypes in patients with myocardial infarction. Adv. Med. Sci. 2019, 64, 224–229. [Google Scholar] [CrossRef]
  73. Wei, F.F.; Zhou, Y.; Thijs, L.; Xue, R.; Dong, B.; He, X.; Liang, W.; Wu, Y.; Jiang, J.; Tan, W.; et al. Visit-to-visit blood pressure variability and clinical outcomes in patients with heart failure with preserved ejection fraction. Hypertension 2021. [Google Scholar] [CrossRef] [PubMed]
  74. Wei, F.F.; Xue, R.; Thijs, L.; Liang, W.; Owusu-Agyeman, M.; He, X.; Staessen, J.A.; Dong, Y.; Liu, C. Associations of left ventricular structure and function with blood pressure in heart failure with preserved ejection fraction: Analysis of the TOPCAT trial. J. Am. Heart Assoc. 2020, 9, e016009. [Google Scholar] [CrossRef] [PubMed]
  75. Bohm, M.; Young, R.; Jhund, P.S.; Solomon, S.D.; Gong, J.; Lefkowitz, M.P.; Rizkala, A.R.; Rouleau, J.L.; Shi, V.C.; Swedberg, K.; et al. Systolic blood pressure, cardiovascular outcomes and efficacy and safety of sacubitril/valsartan (LCZ696) in patients with chronic heart failure and reduced ejection fraction: Results from PARADIGM-HF. Eur. Heart J. 2017, 38, 1132–1143. [Google Scholar] [CrossRef]
  76. Tsujimoto, T.; Kajio, H. Low diastolic blood pressure and adverse outcomes in heart failure with preserved ejection fraction. Int. J. Cardiol. 2018, 263, 69–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Gotte, J.; Zittermann, A.; Deutsch, M.A.; Schramm, R.; Bleiziffer, S.; Hata, M.; Gummert, J.F. Daytime variation in aortic valve surgery and clinical outcome: A propensity score-matched analysis. Ann. Thorac. Surg. 2020, 110, 558–566. [Google Scholar] [CrossRef]
  78. Kenney, P.S.; Nielsen, P.H.; Modrau, I.S. Daytime-dependent cardioprotection in cardiac surgery: A large propensity-matched cohort study. Ann. Thorac. Surg. 2020, 110, 1629–1636. [Google Scholar] [CrossRef]
  79. Michaud, M.; Beland, V.; Noiseux, N.; Forcillo, J.; Stevens, L.M. Daytime variation of clinical outcome in cardiac surgery: A propensity-matched cohort study. J. Cardiothorac. Vasc. Anesth. 2021. [Google Scholar] [CrossRef]
  80. Montaigne, D.; Marechal, X.; Modine, T.; Coisne, A.; Mouton, S.; Fayad, G.; Ninni, S.; Klein, C.; Ortmans, S.; Seunes, C.; et al. Daytime variation of perioperative myocardial injury in cardiac surgery and its prevention by Rev-Erbalpha antagonism: A single-centre propensity-matched cohort study and a randomised study. Lancet 2018, 391, 59–69. [Google Scholar] [CrossRef]
  81. du Fay de Lavallaz, J.; Puelacher, C.; Buse, G.L.; Bolliger, D.; Germanier, D.; Hidvegi, R.; Walter, J.E.; Twerenbold, R.; Strebel, I.; Badertscher, P.; et al. Daytime variation of perioperative myocardial injury in non-cardiac surgery and effect on outcome. Heart 2019, 105, 826–833. [Google Scholar] [CrossRef]
  82. Voulgaris, A.; Marrone, O.; Bonsignore, M.R.; Steiropoulos, P. Chronic kidney disease in patients with obstructive sleep apnea. A narrative review. Sleep Med. Rev. 2019, 47, 74–89. [Google Scholar] [CrossRef] [PubMed]
  83. Rucker, D.; Tonelli, M. Cardiovascular risk and management in chronic kidney disease. Nat. Rev. Nephrol. 2009, 5, 287–296. [Google Scholar] [CrossRef]
  84. Agarwal, R.; Andersen, M.J. Prognostic importance of ambulatory blood pressure recordings in patients with chronic kidney disease. Kidney Int. 2006, 69, 1175–1180. [Google Scholar] [CrossRef] [Green Version]
  85. Davidson, M.B.; Hix, J.K.; Vidt, D.G.; Brotman, D.J. Association of impaired diurnal blood pressure variation with a subsequent decline in glomerular filtration rate. Arch. Intern. Med. 2006, 166, 846–852. [Google Scholar] [CrossRef] [Green Version]
  86. Elung-Jensen, T.; Strandgaard, S.; Kamper, A.L. Longitudinal observations on circadian blood pressure variation in chronic kidney disease stages 3-5. Nephrol. Dial. Transplant. 2008, 23, 2873–2878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Agarwal, R.; Kariyanna, S.S.; Light, R.P. Circadian blood pressure classification scheme and the health of patients with chronic kidney disease. Am. J. Nephrol. 2009, 30, 536–546. [Google Scholar] [CrossRef]
  88. Agarwal, R.; Kariyanna, S.S.; Light, R.P. Prognostic value of circadian blood pressure variation in chronic kidney disease. Am. J. Nephrol. 2009, 30, 547–553. [Google Scholar] [CrossRef]
  89. Fukuda, M.; Munemura, M.; Usami, T.; Nakao, N.; Takeuchi, O.; Kamiya, Y.; Yoshida, A.; Kimura, G. Nocturnal blood pressure is elevated with natriuresis and proteinuria as renal function deteriorates in nephropathy. Kidney Int. 2004, 65, 621–625. [Google Scholar] [CrossRef] [Green Version]
  90. Hussein, A.M.; Malek, H.A.; Saad, M.A. Renoprotective effects of aliskiren on adenine-induced tubulointerstitial nephropathy: Possible underlying mechanisms. Can. J. Physiol. Pharmacol. 2016, 94, 829–837. [Google Scholar] [CrossRef] [Green Version]
  91. Motohashi, H.; Tahara, Y.; Whittaker, D.S.; Wang, H.B.; Yamaji, T.; Wakui, H.; Haraguchi, A.; Yamazaki, M.; Miyakawa, H.; Hama, K.; et al. The circadian clock is disrupted in mice with adenine-induced tubulointerstitial nephropathy. Kidney Int. 2020, 97, 728–740. [Google Scholar] [CrossRef] [Green Version]
  92. Zhang, J.; Liu, C.; Liang, Q.; Zheng, F.; Guan, Y.; Yang, G.; Chen, L. Postnatal deletion of Bmal1 in mice protects against obstructive renal fibrosis via suppressing Gli2 transcription. FASEB J. 2021, 35, e21530. [Google Scholar] [CrossRef]
  93. Rahman, A.; Hasan, A.U.; Nishiyama, A.; Kobori, H. Altered circadian timing system-mediated non-dipping pattern of blood pressure and associated cardiovascular disorders in metabolic and kidney diseases. Int. J. Mol. Sci. 2018, 19, 400. [Google Scholar] [CrossRef] [Green Version]
  94. Otsuka, K.; Cornelissen, G.; Halberg, F.; Oehlerts, G. Excessive circadian amplitude of blood pressure increases risk of ischaemic stroke and nephropathy. J. Med. Eng. Technol. 1997, 21, 23–30. [Google Scholar] [CrossRef]
  95. Velasquez, M.T.; Beddhu, S.; Nobakht, E.; Rahman, M.; Raj, D.S. Ambulatory blood pressure in chronic kidney disease: Ready for prime time? Kidney Int. Rep. 2016, 1, 94–104. [Google Scholar] [CrossRef] [Green Version]
  96. Kuroda, T.; Kario, K.; Hoshide, S.; Hashimoto, T.; Nomura, Y.; Saito, Y.; Mito, H.; Shimada, K. Effects of bedtime vs. morning administration of the long-acting lipophilic angiotensin-converting enzyme inhibitor trandolapril on morning blood pressure in hypertensive patients. Hypertens. Res. 2004, 27, 15–20. [Google Scholar] [CrossRef] [Green Version]
  97. Middeke, M.; Kluglich, M.; Holzgreve, H. Chronopharmacology of captopril plus hydrochlorothiazide in hypertension: Morning versus evening dosing. Chronobiol. Int. 1991, 8, 506–510. [Google Scholar] [CrossRef]
  98. Hermida, R.C.; Ayala, D.E.; Fontao, M.J.; Mojon, A.; Alonso, I.; Fernandez, J.R. Administration-time-dependent effects of spirapril on ambulatory blood pressure in uncomplicated essential hypertension. Chronobiol. Int. 2010, 27, 560–574. [Google Scholar] [CrossRef]
  99. Witte, K.; Weisser, K.; Neubeck, M.; Mutschler, E.; Lehmann, K.; Hopf, R.; Lemmer, B. Cardiovascular effects, pharmacokinetics, and converting enzyme inhibition of enalapril after morning versus evening administration. Clin. Pharmacol. Ther. 1993, 54, 177–186. [Google Scholar] [CrossRef]
  100. Macchiarulo, C.; Pieri, R.; Mitolo, D.C.; Pirrelli, A. Management of antihypertensive treatment with Lisinopril: A chronotherapeutic approach. Eur. Rev. Med. Pharmacol. Sci. 1999, 3, 269–275. [Google Scholar]
  101. Palatini, P.; Racioppa, A.; Raule, G.; Zaninotto, M.; Penzo, M.; Pessina, A.C. Effect of timing of administration on the plasma ACE inhibitory activity and the antihypertensive effect of quinapril. Clin. Pharmacol. Ther. 1992, 52, 378–383. [Google Scholar] [CrossRef]
  102. Palatini, P.; Mos, L.; Motolese, M.; Mormino, P.; Del Torre, M.; Varotto, L.; Pavan, E.; Pessina, A.C. Effect of evening versus morning benazepril on 24-hour blood pressure: A comparative study with continuous intraarterial monitoring. Int. J. Clin. Pharmacol. Ther. Toxicol. 1993, 31, 295–300. [Google Scholar]
  103. Zaslavskaia, R.M.; Narmanova, O.Z.; Teiblium, M.M.; Kalimurzina, B.S. Time-dependent effects of ramipril in patients with hypertension of 2 stage. Klin. Meditsina 1999, 77, 41–44. [Google Scholar]
  104. Kohno, I.; Ijiri, H.; Takusagawa, M.; Yin, D.F.; Sano, S.; Ishihara, T.; Sawanobori, T.; Komori, S.; Tamura, K. Effect of imidapril in dipper and nondipper hypertensive patients: Comparison between morning and evening administration. Chronobiol. Int. 2000, 17, 209–219. [Google Scholar] [CrossRef] [PubMed]
  105. Hermida, R.C.; Calvo, C.; Ayala, D.E.; Dominguez, M.J.; Covelo, M.; Fernandez, J.R.; Mojon, A.; Lopez, J.E. Administration time-dependent effects of valsartan on ambulatory blood pressure in hypertensive subjects. Hypertension 2003, 42, 283–290. [Google Scholar] [CrossRef] [Green Version]
  106. Hermida, R.C.; Calvo, C.; Ayala, D.E.; Fernandez, J.R.; Covelo, M.; Mojon, A.; Lopez, J.E. Treatment of non-dipper hypertension with bedtime administration of valsartan. J. Hypertens. 2005, 23, 1913–1922. [Google Scholar] [CrossRef]
  107. Hermida, R.C.; Ayala, D.E.; Fontao, M.J.; Mojon, A.; Fernandez, J.R. Chronotherapy with valsartan/amlodipine fixed combination: Improved blood pressure control of essential hypertension with bedtime dosing. Chronobiol. Int. 2010, 27, 1287–1303. [Google Scholar] [CrossRef]
  108. Hermida, R.C.; Ayala, D.E.; Fernandez, J.R.; Calvo, C. Comparison of the efficacy of morning versus evening administration of telmisartan in essential hypertension. Hypertension 2007, 50, 715–722. [Google Scholar] [CrossRef]
  109. Fukuda, M.; Yamanaka, T.; Mizuno, M.; Motokawa, M.; Shirasawa, Y.; Miyagi, S.; Nishio, T.; Yoshida, A.; Kimura, G. Angiotensin II type 1 receptor blocker, olmesartan, restores nocturnal blood pressure decline by enhancing daytime natriuresis. J. Hypertens. 2008, 26, 583–588. [Google Scholar] [CrossRef] [PubMed]
  110. Bankir, L.; Bochud, M.; Maillard, M.; Bovet, P.; Gabriel, A.; Burnier, M. Nighttime blood pressure and nocturnal dipping are associated with daytime urinary sodium excretion in African subjects. Hypertension 2008, 51, 891–898. [Google Scholar] [CrossRef] [Green Version]
  111. Del Giorno, R.; Troiani, C.; Gabutti, S.; Stefanelli, K.; Puggelli, S.; Gabutti, L. Impaired daytime urinary sodium excretion impacts nighttime blood pressure and nocturnal dipping at older ages in the general population. Nutrients 2020, 12, 2013. [Google Scholar] [CrossRef]
  112. Hermida, R.C.; Ayala, D.E.; Chayan, L.; Mojon, A.; Fernandez, J.R. Administration-time-dependent effects of olmesartan on the ambulatory blood pressure of essential hypertension patients. Chronobiol. Int. 2009, 26, 61–79. [Google Scholar] [CrossRef]
  113. Smolensky, M.H.; Hermida, R.C.; Portaluppi, F. Comparison of the efficacy of morning versus evening administration of olmesartan in uncomplicated essential hypertension. Chronobiol. Int. 2007, 24, 171–181. [Google Scholar] [CrossRef]
  114. Hermida, R.C.; Fernandez, J.R.; Ayala, D.E.; Mojon, A.; Iglesias, M. Influence of aspirin usage on blood pressure: Dose and administration-time dependencies. Chronobiol. Int. 1997, 14, 619–637. [Google Scholar] [CrossRef] [PubMed]
  115. Hermida, R.C.; Ayala, D.E.; Iglesias, M.; Mojon, A.; Silva, I.; Ucieda, R.; Fernandez, J.R. Time-dependent effects of low-dose aspirin administration on blood pressure in pregnant women. Hypertension 1997, 30, 589–595. [Google Scholar] [CrossRef]
  116. Hermida, R.C.; Ayala, D.E.; Mojon, A.; Fernandez, J.R. Ambulatory blood pressure control with bedtime aspirin administration in subjects with prehypertension. Am. J. Hypertens. 2009, 22, 896–903. [Google Scholar] [CrossRef]
  117. Hermida, R.C.; Ayala, D.E.; Calvo, C.; Lopez, J.E.; Mojon, A.; Rodriguez, M.; Fernandez, J.R. Differing administration time-dependent effects of aspirin on blood pressure in dipper and non-dipper hypertensives. Hypertension 2005, 46, 1060–1068. [Google Scholar] [CrossRef] [Green Version]
  118. Hermida, R.C.; Ayala, D.E.; Calvo, C.; Lopez, J.E. Aspirin administered at bedtime, but not on awakening, has an effect on ambulatory blood pressure in hypertensive patients. J. Am. Coll. Cardiol. 2005, 46, 975–983. [Google Scholar] [CrossRef]
  119. Chen, L.; Yang, G.; Zhang, J.; Ren, B.; Tang, S.; Li, X.; FitzGerald, G.A. Time-Dependent Hypotensive Effect of Aspirin in Mice. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 2819–2826. [Google Scholar] [CrossRef] [Green Version]
  120. Hermida, R.C.; Ayala, D.E.; Mojon, A.; Fernandez, J.R. Chronotherapy with nifedipine GITS in hypertensive patients: Improved efficacy and safety with bedtime dosing. Am. J. Hypertens. 2008, 21, 948–954. [Google Scholar] [CrossRef] [Green Version]
  121. Hermida, R.C.; Calvo, C.; Ayala, D.E.; Lopez, J.E.; Rodriguez, M.; Chayan, L.; Mojon, A.; Fontao, M.J.; Fernandez, J.R. Dose- and administration time-dependent effects of nifedipine gits on ambulatory blood pressure in hypertensive subjects. Chronobiol. Int. 2007, 24, 471–493. [Google Scholar] [CrossRef] [PubMed]
  122. Hermida, R.C.; Ayala, D.E.; Mojon, A.; Alonso, I.; Fernandez, J.R. Reduction of morning blood pressure surge after treatment with nifedipine GITS at bedtime, but not upon awakening, in essential hypertension. Blood Press. Monit. 2009, 14, 152–159. [Google Scholar] [CrossRef]
  123. Hermida, R.C.; Crespo, J.J.; Dominguez-Sardina, M.; Otero, A.; Moya, A.; Rios, M.T.; Sineiro, E.; Castineira, M.C.; Callejas, P.A.; Pousa, L.; et al. Bedtime hypertension treatment improves cardiovascular risk reduction: The Hygia Chronotherapy Trial. Eur. Heart J. 2020, 41, 4565–4576. [Google Scholar] [CrossRef] [PubMed]
  124. Kreutz, R.; Kjeldsen, S.E.; Burnier, M.; Narkiewicz, K.; Oparil, S.; Mancia, G. Disregard the reported data from the HYGIA project: Blood pressure medication not to be routinely dosed at bedtime. J. Hypertens. 2020, 38, 2144–2145. [Google Scholar] [CrossRef] [PubMed]
  125. Rahman, M.; Greene, T.; Phillips, R.A.; Agodoa, L.Y.; Bakris, G.L.; Charleston, J.; Contreras, G.; Gabbai, F.; Hiremath, L.; Jamerson, K.; et al. A trial of 2 strategies to reduce nocturnal blood pressure in blacks with chronic kidney disease. Hypertension 2013, 61, 82–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Tamura, K.; Kohno, I.; Yuzo, S.; Wakasugi, K.; Achiwa, S.; Imanishi, Y.; Cugini, P.; Halberg, F. Antihypertensive individualized therapeutic strategy. Dif. Soc. 1991, 6, 109–124. [Google Scholar]
  127. Watanabe, Y.; Halberg, F.; Otsuka, K.; Cornelissen, G. Toward a personalized chronotherapy of high blood pressure and a circadian overswing. Clin. Exp. Hypertens. 2013, 35, 257–266. [Google Scholar] [CrossRef]
Figure 1. Molecular mechanism of circadian clock in humans. The molecular oscillator comprises interconnected transcription-translation feedback loops. The expression of CCGs is driven by the output of the molecular clock and adjusts circadian clock of cardiovascular and renal function. ROR activates and REV-ERB represses RORE-mediated transcription. PPARα and PPARγ regulate the expression of BMAL1 and REV-ERB via binding to PPRE in their promoters. As the PPAR partner, RXR inhibits the transcriptional activity of BMAL1-CLOCK complex. RORE, ROR response element. PPRE, PPAR response element. RXR, retinoid X receptor.
Figure 1. Molecular mechanism of circadian clock in humans. The molecular oscillator comprises interconnected transcription-translation feedback loops. The expression of CCGs is driven by the output of the molecular clock and adjusts circadian clock of cardiovascular and renal function. ROR activates and REV-ERB represses RORE-mediated transcription. PPARα and PPARγ regulate the expression of BMAL1 and REV-ERB via binding to PPRE in their promoters. As the PPAR partner, RXR inhibits the transcriptional activity of BMAL1-CLOCK complex. RORE, ROR response element. PPRE, PPAR response element. RXR, retinoid X receptor.
Biomolecules 11 00868 g001
Table 1. The effect of the core clock gene manipulation on BP phenotypes in rodents.
Table 1. The effect of the core clock gene manipulation on BP phenotypes in rodents.
Gene EditingSpeciesPositionPhenotype of BPCircadian Rhythmof BPReference
Bmal1 KOmouseglobalNo[33,39]
Bmal1 KOratglobalYes[40]
Clock KOmouseglobalYes[41]
Clock mutantmouseglobalDay-night variation↓Yes[42]
Per1 KO (129/sv)mouseglobalYes[43]
Per1 KO (C57BL/6)mouseglobalYes[44]
Per2 KOmouseglobal=Yes[45]
Per2 mutantmouseglobalDBP↓Yes[46]
Cry1/2 KOmouseglobalSalt-sensitive hypertensionNo[47]
Bmal1 KOmousesmooth muscleYes[48]
Bmal1 KOmousebrown adipocytesNo[49]
Bmal1 KOmouseendothelial cellsYes[50]
Bmal1 KOmousetubular cellsSBP↓Yes[51]
Bmal1 KOmouserenin-secreting cellsYes[52]
Bmal1 KOmousecollecting ductYes[53]
Bmal1 KOmousethick ascending limb, distal convoluted tubule, and collecting ductYes[54]
Phenotype and circadian rhythm of BP compared to control mice under 12:12 L:D cycles. ↓ means decline; ↑ means increase; = means no difference; SBP, systolic BP; DBP, diastolic BP.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhang, J.; Sun, R.; Jiang, T.; Yang, G.; Chen, L. Circadian Blood Pressure Rhythm in Cardiovascular and Renal Health and Disease. Biomolecules 2021, 11, 868. https://doi.org/10.3390/biom11060868

AMA Style

Zhang J, Sun R, Jiang T, Yang G, Chen L. Circadian Blood Pressure Rhythm in Cardiovascular and Renal Health and Disease. Biomolecules. 2021; 11(6):868. https://doi.org/10.3390/biom11060868

Chicago/Turabian Style

Zhang, Jiayang, Ruoyu Sun, Tingting Jiang, Guangrui Yang, and Lihong Chen. 2021. "Circadian Blood Pressure Rhythm in Cardiovascular and Renal Health and Disease" Biomolecules 11, no. 6: 868. https://doi.org/10.3390/biom11060868

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop