1
|
Costello HM, Eikenberry SA, Cheng KY, Broderick B, Joshi AS, Scott GR, McKee A, Mendez VM, Douma LG, Crislip GR, Gumz ML. Sex differences in the adrenal circadian clock: a role for BMAL1 in the regulation of urinary aldosterone excretion and renal electrolyte balance in mice. Am J Physiol Renal Physiol 2025; 328:F1-F14. [PMID: 39447118 DOI: 10.1152/ajprenal.00177.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 10/26/2024] Open
Abstract
Brain and muscle ARNT-Like 1 (BMAL1) is a circadian clock transcription factor that regulates physiological functions. Male adrenal-specific Bmal1 (ASCre/+::Bmal1) KO mice displayed blunted serum corticosterone rhythms, altered blood pressure rhythm, and altered timing of eating, but there is a lack of knowledge in females. This study investigates the role of adrenal BMAL1 in renal electrolyte handling and urinary aldosterone levels in response to low salt in male and female mice. Mice were placed in metabolic cages to measure 12-h urinary aldosterone after a standard diet and 7 days low-salt diet, as well as daily body weight, 12-h food and water intake, and renal sodium and potassium balance. Adrenal glands and kidneys were collected at ZT0 or ZT12 to measure the expression of aldosterone synthesis genes and clock genes. Compared with littermate controls, ASCre/+::Bmal1 KO male and female mice displayed increased urinary aldosterone in response to a low-salt diet, although mRNA expression of aldosterone synthesis genes was decreased. Timing of food intake was altered in ASCre/+::Bmal1 KO male and female mice, with a blunted night/day ratio. ASCre/+::Bmal1 KO female mice displayed decreases in renal sodium excretion in response to low salt, but both male and female KO mice had changes in sodium balance that were time-of-day-dependent. In addition, sex differences were found in adrenal and kidney clock gene expression. Notably, this study highlights sex differences in clock gene expression that could contribute to sex differences in physiological functions.NEW & NOTEWORTHY Our findings highlight the importance of sex as well as time-of-day in understanding the role of the circadian clock in the regulation of homeostasis. Time-of-day is a key biological variable that is often ignored in research, particularly in preclinical rodent studies. Our findings demonstrate important differences in several measures at 6 AM compared with 6 PM. Consideration of time-of-day is critical for the translation of findings in nocturnal rodent physiology to diurnal human physiology.
Collapse
Affiliation(s)
- Hannah M Costello
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
| | - Sophia A Eikenberry
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
| | - Kit-Yan Cheng
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
| | - Bryanna Broderick
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Advay S Joshi
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Gianna R Scott
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Annalisse McKee
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Victor M Mendez
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Lauren G Douma
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, United States
| | - G Ryan Crislip
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
| | - Michelle L Gumz
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, United States
- Research, North Florida/South Georgia Malcolm Randall Veterans Affairs Medical Center, Gainesville, Florida, United States
| |
Collapse
|
2
|
Visniauskas B, Ogola BO, Kilanowski-Doroh I, Harris NR, Diaz ZT, Horton AC, Blessinger SA, McNally AB, Zimmerman MA, Arnold AC, Lindsey SH. Hypertension disrupts the vascular clock in both sexes. Am J Physiol Heart Circ Physiol 2024; 327:H765-H777. [PMID: 39058434 PMCID: PMC11649248 DOI: 10.1152/ajpheart.00131.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Blood pressure (BP) displays a circadian rhythm and disruptions in this pattern elevate cardiovascular risk. Although both central and peripheral clock genes are implicated in these processes, the importance of vascular clock genes is not fully understood. BP, vascular reactivity, and the renin-angiotensin-aldosterone system display overt sex differences, but whether changes in circadian patterns underlie these differences is unknown. Therefore, we hypothesized that circadian rhythms and vascular clock genes would differ across sex and would be blunted by angiotensin II (ANG II)-induced hypertension. ANG II infusion elevated BP and disrupted circadian patterns similarly in both males and females. In females, an impact on heart rate (HR) and locomotor activity was revealed, whereas in males hypertension suppressed baroreflex sensitivity (BRS). A marked disruption in the vascular expression patterns of period circadian regulator 1 (Per1) and brain and muscle aryl hydrocarbon receptor nuclear translocator like protein 1 (Bmal1) was noted in both sexes. Vascular expression of the G protein-coupled estrogen receptor (Gper1) also showed diurnal synchronization in both sexes that was similar to that of Per1 and Per2 and disrupted by hypertension. In contrast, vascular expression of estrogen receptor 1 (Esr1) showed a diurnal rhythm and hypertension-induced disruption only in females. This study shows a strikingly similar impact of hypertension on BP rhythmicity, vascular clock genes, and vascular estrogen receptor expression in both sexes. We identified a greater impact of hypertension on locomotor activity and heart rate in females and on baroreflex sensitivity in males and also revealed a diurnal regulation of vascular estrogen receptors. These insights highlight the intricate ties between circadian biology, sex differences, and cardiovascular regulation.NEW & NOTEWORTHY This study reveals that ANG II-induced hypertension disrupts the circadian rhythm of blood pressure in both male and female mice, with parallel effects on vascular clock gene and estrogen receptor diurnal patterns. Notably, sex-specific responses to hypertension in terms of locomotor activity, heart rate, and baroreflex sensitivity are revealed. These findings pave the way for chronotherapeutic strategies tailored to mitigate cardiovascular risks associated with disrupted circadian rhythms in hypertension.
Collapse
Affiliation(s)
- Bruna Visniauskas
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Isabella Kilanowski-Doroh
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Nicholas R Harris
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Zaidmara T Diaz
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Alec C Horton
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Sophia A Blessinger
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Alexandra B McNally
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Margaret A Zimmerman
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Amy C Arnold
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Tulane Center of Excellence in Sex-Based Biology and Medicine, Tulane University, New Orleans, Louisiana, United States
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana, United States
| |
Collapse
|
3
|
Juffre A, Gumz ML. Recent advances in understanding the kidney circadian clock mechanism. Am J Physiol Renal Physiol 2024; 326:F382-F393. [PMID: 38174377 PMCID: PMC11207534 DOI: 10.1152/ajprenal.00214.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024] Open
Abstract
Circadian rhythms are endogenous biological oscillations that regulate various physiological processes in organisms, including kidney function. The kidney plays a vital role in maintaining homeostasis by regulating water and electrolyte balance, blood pressure, and excretion of metabolic waste products, all of which display circadian rhythmicity. For this reason, studying the circadian regulation of the kidney is important, and the time of day is a biological and experimental variable that must be considered. Over the past decade, considerable progress has been made in understanding the molecular mechanisms underlying circadian regulation within the kidney. In this review, the current knowledge regarding circadian rhythms in the kidney is explored, focusing on the molecular clock machinery, circadian control of renal functions, and the impact of disrupted circadian rhythms on kidney health. In addition, parameters that should be considered and future directions are outlined in this review.
Collapse
Affiliation(s)
- Alexandria Juffre
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
| | - Michelle L Gumz
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
4
|
Benjamin JI, Pollock DM. Current perspective on circadian function of the kidney. Am J Physiol Renal Physiol 2024; 326:F438-F459. [PMID: 38134232 PMCID: PMC11207578 DOI: 10.1152/ajprenal.00247.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/28/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023] Open
Abstract
Behavior and function of living systems are synchronized by the 24-h rotation of the Earth that guides physiology according to time of day. However, when behavior becomes misaligned from the light-dark cycle, such as in rotating shift work, jet lag, and even unusual eating patterns, adverse health consequences such as cardiovascular or cardiometabolic disease can arise. The discovery of cell-autonomous molecular clocks expanded interest in regulatory systems that control circadian physiology including within the kidney, where function varies along a 24-h cycle. Our understanding of the mechanisms for circadian control of physiology is in the early stages, and so the present review provides an overview of what is known and the many gaps in our current understanding. We include a particular focus on the impact of eating behaviors, especially meal timing. A better understanding of the mechanisms guiding circadian function of the kidney is expected to reveal new insights into causes and consequences of a wide range of disorders involving the kidney, including hypertension, obesity, and chronic kidney disease.
Collapse
Affiliation(s)
- Jazmine I Benjamin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - David M Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
5
|
Shafer BM, Kogan SA, McHill AW. Pressure Building Against the Clock: The Impact of Circadian Misalignment on Blood Pressure. Curr Hypertens Rep 2024; 26:31-42. [PMID: 37837518 PMCID: PMC10916535 DOI: 10.1007/s11906-023-01274-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2023] [Indexed: 10/16/2023]
Abstract
PURPOSE OF REVIEW Misalignment between the endogenous biological timing system and behavioral activities (i.e., sleep/wake, eating, activity) contributes to adverse cardiovascular health. In this review, we discuss the effects of recurring circadian misalignment on blood pressure regulation and the implications for hypertension development. Additionally, we highlight emerging therapeutic approaches designed to mitigate the negative cardiovascular consequences elicited by circadian disruption. RECENT FINDINGS Circadian misalignment elicited by work schedules that require individuals to be awake during the biological night (i.e., shift work) alters 24-h blood pressure rhythms. Mechanistically, circadian misalignment appears to alter blood pressure via changes in autonomic nervous system balance, variations to sodium retention, dysregulation of endothelial vasodilatory responsiveness, and activation of proinflammatory mechanisms. Recurring circadian misalignment produced by a mismatch in sleep timing on free days vs. work days (i.e., social jetlag) appears to have no direct effects on prevailing blood pressure levels in healthy adults; though, circadian disruptions resulting from social jetlag may increase the risk of hypertension through enhanced sympathetic activation and/or obesity. Furthermore, social jetlag assessment may be a useful metric in shift work populations where the magnitude of circadian misalignment may be greater than in the general population. Circadian misalignment promotes unfavorable changes to 24-h blood pressure rhythms, most notably in shift working populations. While light therapy, melatonin supplementation, and the timing of drug administration may improve cardiovascular outcomes, interventions designed to target the effects of circadian misalignment on blood pressure regulation are warranted.
Collapse
Affiliation(s)
- Brooke M Shafer
- Sleep, Chronobiology, and Health Laboratory, School of Nursing, Oregon Health & Science University, 3455 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Sophia A Kogan
- Sleep, Chronobiology, and Health Laboratory, School of Nursing, Oregon Health & Science University, 3455 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Andrew W McHill
- Sleep, Chronobiology, and Health Laboratory, School of Nursing, Oregon Health & Science University, 3455 SW US Veterans Hospital Rd, Portland, OR, 97239, USA.
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
6
|
Costello HM, Sharma RK, McKee AR, Gumz ML. Circadian Disruption and the Molecular Clock in Atherosclerosis and Hypertension. Can J Cardiol 2023; 39:1757-1771. [PMID: 37355229 PMCID: PMC11446228 DOI: 10.1016/j.cjca.2023.06.416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/05/2023] [Accepted: 06/18/2023] [Indexed: 06/26/2023] Open
Abstract
Circadian rhythms are crucial for maintaining vascular function and disruption of these rhythms are associated with negative health outcomes including cardiovascular disease and hypertension. Circadian rhythms are regulated by the central clock within the suprachiasmatic nucleus of the hypothalamus and peripheral clocks located in nearly every cell type in the body, including cells within the heart and vasculature. In this review, we summarize the most recent preclinical and clinical research linking circadian disruption, with a focus on molecular circadian clock mechanisms, in atherosclerosis and hypertension. Furthermore, we provide insight into potential future chronotherapeutics for hypertension and vascular disease. A better understanding of the influence of daily rhythms in behaviour, such as sleep/wake cycles, feeding, and physical activity, as well as the endogenous circadian system on cardiovascular risk will help pave the way for targeted approaches in atherosclerosis and hypertension treatment/prevention.
Collapse
Affiliation(s)
- Hannah M Costello
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, USA; Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA; Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, Florida, USA.
| | - Ravindra K Sharma
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, USA; Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA; Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, Florida, USA
| | - Annalisse R McKee
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, USA; Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Michelle L Gumz
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, USA; Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA; Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, Florida, USA; Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
7
|
Buncha V, Cherezova A, Alexander S, Baranovskaya I, Coleman KA, Cherian-Shaw M, Brands MW, Sullivan JC, O'Connor PM, Mamenko M. Aldosterone Antagonism Is More Effective at Reducing Blood Pressure and Excessive Renal ENaC Activity in AngII-Infused Female Rats Than in Males. Hypertension 2023; 80:2196-2208. [PMID: 37593894 PMCID: PMC10528186 DOI: 10.1161/hypertensionaha.123.21287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND AngII (angiotensin II)-dependent hypertension causes comparable elevations of blood pressure (BP), aldosterone levels, and renal ENaC (epithelial Na+ channel) activity in male and female rodents. Mineralocorticoid receptor (MR) antagonism has a limited antihypertensive effect associated with insufficient suppression of renal ENaC in male rodents with AngII-hypertension. While MR blockade effectively reduces BP in female mice with salt-sensitive and leptin-induced hypertension, MR antagonism has not been studied in female rodents with AngII-hypertension. We hypothesize that overstimulation of renal MR signaling drives redundant ENaC-mediated Na+ reabsorption and BP increase in female rats with AngII-hypertension. METHODS We employ a combination of physiological, pharmacological, biochemical, and biophysical approaches to compare the effect of MR inhibitors on BP and ENaC activity in AngII-infused male and female Sprague Dawley rats. RESULTS MR blockade markedly attenuates AngII-hypertension in female rats but has only a marginal effect in males. Spironolactone increases urinary sodium excretion and urinary sodium-to-potassium ratio in AngII-infused female, but not male, rats. The expression of renal MR and HSD11β2 (11β-hydroxysteroid dehydrogenase type 2) that determines the availability of MR to aldosterone is significantly higher in AngII-infused female rats than in males. ENaC activity is ≈2× lower in spironolactone-treated AngII-infused female rats than in males. Reduced ENaC activity in AngII-infused female rats on spironolactone correlates with increased interaction with ubiquitin ligase Nedd4-2 (neural precursor cell expressed developmentally down-regulated protein 4-2), targeting ENaC for degradation. CONCLUSIONS MR-ENaC axis is the primary determinant of excessive renal sodium reabsorption and an attractive antihypertensive target in female rats with AngII-hypertension, but not in males.
Collapse
Affiliation(s)
- Vadym Buncha
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Alena Cherezova
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Sati Alexander
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Irina Baranovskaya
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Kathleen A Coleman
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Mary Cherian-Shaw
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Michael W Brands
- Department of Physiology, Medical College of Georgia, Augusta University
| | | | - Paul M O'Connor
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Mykola Mamenko
- Department of Physiology, Medical College of Georgia, Augusta University
| |
Collapse
|
8
|
Rios FJ, Montezano AC, Camargo LL, Touyz RM. Impact of Environmental Factors on Hypertension and Associated Cardiovascular Disease. Can J Cardiol 2023; 39:1229-1243. [PMID: 37422258 DOI: 10.1016/j.cjca.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/24/2023] [Accepted: 07/02/2023] [Indexed: 07/10/2023] Open
Abstract
Hypertension is the primary cause of cardiovascular diseases and is responsible for nearly 9 million deaths worldwide annually. Increasing evidence indicates that in addition to pathophysiologic processes, numerous environmental factors, such as geographic location, lifestyle choices, socioeconomic status, and cultural practices, influence the risk, progression, and severity of hypertension, even in the absence of genetic risk factors. In this review, we discuss the impact of some environmental determinants on hypertension. We focus on clinical data from large population studies and discuss some potential molecular and cellular mechanisms. We highlight how these environmental determinants are interconnected, as small changes in one factor might affect others, and further affect cardiovascular health. In addition, we discuss the crucial impact of socioeconomic factors and how these determinants influence diverse communities with economic disparities. Finally, we address opportunities and challenges for new research to address gaps in knowledge on understanding molecular mechanisms whereby environmental factors influence development of hypertension and associated cardiovascular disease.
Collapse
Affiliation(s)
- Francisco J Rios
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| | - Augusto C Montezano
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Livia L Camargo
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
9
|
Anderson ST, Meng H, Brooks TG, Tang SY, Lordan R, Sengupta A, Nayak S, Mřela A, Sarantopoulou D, Lahens NF, Weljie A, Grant GR, Bushman FD, FitzGerald GA. Sexual dimorphism in the response to chronic circadian misalignment on a high-fat diet. Sci Transl Med 2023; 15:eabo2022. [PMID: 37196066 DOI: 10.1126/scitranslmed.abo2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/14/2023] [Indexed: 05/19/2023]
Abstract
Longitudinal studies associate shiftwork with cardiometabolic disorders but do not establish causation or elucidate mechanisms of disease. We developed a mouse model based on shiftwork schedules to study circadian misalignment in both sexes. Behavioral and transcriptional rhythmicity were preserved in female mice despite exposure to misalignment. Females were protected from the cardiometabolic impact of circadian misalignment on a high-fat diet seen in males. The liver transcriptome and proteome revealed discordant pathway perturbations between the sexes. Tissue-level changes were accompanied by gut microbiome dysbiosis only in male mice, biasing toward increased potential for diabetogenic branched chain amino acid production. Antibiotic ablation of the gut microbiota diminished the impact of misalignment. In the United Kingdom Biobank, females showed stronger circadian rhythmicity in activity and a lower incidence of metabolic syndrome than males among job-matched shiftworkers. Thus, we show that female mice are more resilient than males to chronic circadian misalignment and that these differences are conserved in humans.
Collapse
Affiliation(s)
- Seán T Anderson
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hu Meng
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas G Brooks
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Soon Yew Tang
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arjun Sengupta
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Soumyashant Nayak
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Antonijo Mřela
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dimitra Sarantopoulou
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas F Lahens
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aalim Weljie
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gregory R Grant
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Frederic D Bushman
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Garret A FitzGerald
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Zietara A, Spires DR, Juffre A, Costello HM, Crislip GR, Douma LG, Levchenko V, Dissanayake LV, Klemens CA, Nikolaienko O, Geurts AM, Gumz ML, Staruschenko A. Knockout of the Circadian Clock Protein PER1 (Period1) Exacerbates Hypertension and Increases Kidney Injury in Dahl Salt-Sensitive Rats. Hypertension 2022; 79:2519-2529. [PMID: 36093781 PMCID: PMC9669134 DOI: 10.1161/hypertensionaha.122.19316] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/14/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Circadian rhythms play an essential role in physiological function. The molecular clock that underlies circadian physiological function consists of a core group of transcription factors, including the protein PER1 (Period1). Studies in mice show that PER1 plays a role in the regulation of blood pressure and renal sodium handling; however, the results are dependent on the strain being studied. Using male Dahl salt-sensitive (SS) rats with global knockout of PER1 (SSPer1-/-), we aim to test the hypothesis that PER1 plays a key role in the regulation of salt-sensitive blood pressure. METHODS The model was generated using CRISPR/Cas9 and was characterized using radiotelemetry and measures of renal function and circadian rhythm. RESULTS SSPer1-/- rats had similar mean arterial pressure when fed a normal 0.4% NaCl diet but developed augmented hypertension after three weeks on a high-salt (4% NaCl) diet. Despite being maintained on a normal 12:12 light:dark cycle, SSPer1-/- rats exhibited desynchrony mean arterial pressure rhythms on a high-salt diet, as evidenced by increased variability in the time of peak mean arterial pressure. SSPer1-/- rats excrete less sodium after three weeks on the high-salt diet. Furthermore, SSPer1-/- rats exhibited decreased creatinine clearance, a measurement of renal function, as well as increased signs of kidney tissue damage. SSPer1-/- rats also exhibited higher plasma aldosterone levels. CONCLUSIONS Altogether, our findings demonstrate that loss of PER1 in Dahl SS rats causes an array of deleterious effects, including exacerbation of the development of salt-sensitive hypertension and renal damage.
Collapse
Affiliation(s)
- Adrian Zietara
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Denisha R. Spires
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Alexandria Juffre
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
| | - Hannah M. Costello
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
| | - G. Ryan Crislip
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
| | - Lauren G. Douma
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
| | - Vladislav Levchenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
| | - Lashodya V. Dissanayake
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
| | - Christine A. Klemens
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL 33602, USA
| | - Oksana Nikolaienko
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kyiv, Ukraine
| | - Aron M. Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Michelle L. Gumz
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL 32610, USA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL 33602, USA
- James A. Haley Veterans’ Hospital, Tampa, FL 33612, USA
| |
Collapse
|
11
|
rs2253820 Variant Controls Blood Pressure Dip After Stroke by Increasing CLOCK–BMAL1 Expression. Transl Stroke Res 2022:10.1007/s12975-022-01063-y. [DOI: 10.1007/s12975-022-01063-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 10/16/2022]
|
12
|
Patil CN, Ritter ML, Wackman KK, Oliveira V, Balapattabi K, Grobe CC, Brozoski DT, Reho JJ, Nakagawa P, Mouradian GC, Kriegel AJ, Kwitek AE, Hodges MR, Segar JL, Sigmund CD, Grobe JL. Cardiometabolic effects of DOCA-salt in male C57BL/6J mice are variably dependent on sodium and nonsodium components of diet. Am J Physiol Regul Integr Comp Physiol 2022; 322:R467-R485. [PMID: 35348007 PMCID: PMC9054347 DOI: 10.1152/ajpregu.00017.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 01/22/2023]
Abstract
Hypertension characterized by low circulating renin activity accounts for roughly 25%-30% of primary hypertension in humans and can be modeled experimentally via deoxycorticosterone acetate (DOCA)-salt treatment. In this model, phenotypes develop in progressive phases, although the timelines and relative contributions of various mechanisms to phenotype development can be distinct between laboratories. To explore interactions among environmental influences such as diet formulation and dietary sodium (Na) content on phenotype development in the DOCA-salt paradigm, we examined an array of cardiometabolic endpoints in young adult male C57BL/6J mice during sham or DOCA-salt treatments when mice were maintained on several common, commercially available laboratory rodent "chow" diets including PicoLab 5L0D (0.39% Na), Envigo 7913 (0.31% Na), Envigo 2920x (0.15% Na), or a customized version of Envigo 2920x (0.4% Na). Energy balance (weight gain, food intake, digestive efficiency, and energy efficiency), fluid and electrolyte homeostasis (fluid intake, Na intake, fecal Na content, hydration, and fluid compartmentalization), renal functions (urine production rate, glomerular filtration rate, urine Na excretion, renal expression of renin, vasopressin receptors, aquaporin-2 and relationships among markers of vasopressin release, aquaporin-2 shedding, and urine osmolality), and blood pressure, all exhibited changes that were subject to interactions between diet and DOCA-salt. Interestingly, some of these phenotypes, including blood pressure and hydration, were dependent on nonsodium dietary components, as Na-matched diets resulted in distinct phenotype development. These findings provide a broad and robust illustration of an environment × treatment interaction that impacts the use and interpretation of a common rodent model of low-renin hypertension.
Collapse
Affiliation(s)
- Chetan N Patil
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - McKenzie L Ritter
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kelsey K Wackman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Vanessa Oliveira
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Connie C Grobe
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Daniel T Brozoski
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John J Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Gary C Mouradian
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alison J Kriegel
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Anne E Kwitek
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jeffrey L Segar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Curt D Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
13
|
Murray A, Tharmalingam S, Khurana S, Lalonde C, Nguyen P, Tai TC. Effect of Prenatal Glucocorticoid Exposure on Circadian Rhythm Gene Expression in the Brains of Adult Rat Offspring. Cells 2022; 11:cells11101613. [PMID: 35626652 PMCID: PMC9139626 DOI: 10.3390/cells11101613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/03/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
Circadian clocks control many vital aspects of physiology from the sleep-wake cycle to metabolism. The circadian clock operates through transcriptional-translational feedback loops. The normal circadian signaling relies on a ‘master clock’, located in the suprachiasmatic nucleus (SCN), which synchronizes peripheral oscillators. Glucocorticoid receptor (GR) signaling has the ability to reset the phase of peripheral clocks. It has been shown that maternal exposure to glucocorticoids (GCs) can lead to modification of hypothalamic-pituitary-adrenal (HPA) function, impact stress-related behaviors, and result in a hypertensive state via GR activation. We previously demonstrated altered circadian rhythm signaling in the adrenal glands of offspring exposed to the synthetic GC, dexamethasone (Dex). Results from the current study show that prenatal exposure to Dex affects circadian rhythm gene expression in a brain region-specific and a sex-specific manner within molecular oscillators of the amygdala, hippocampus, paraventricular nucleus, and prefrontal cortex, as well as the main oscillator in the SCN. Results also show that spontaneously hypertensive rats (SHR) exhibited dysregulated circadian rhythm gene expression in these same brain regions compared with normotensive Wistar-Kyoto rats (WKY), although the pattern of dysregulation was markedly different from that seen in adult offspring prenatally exposed to GCs.
Collapse
Affiliation(s)
- Alyssa Murray
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | - Sujeenthar Tharmalingam
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Sandhya Khurana
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
| | - Christine Lalonde
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Phong Nguyen
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | - T. C. Tai
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Correspondence: ; Tel.: +1-705-662-7239
| |
Collapse
|
14
|
Ibarz-Blanch N, Morales D, Calvo E, Ros-Medina L, Muguerza B, Bravo FI, Suárez M. Role of Chrononutrition in the Antihypertensive Effects of Natural Bioactive Compounds. Nutrients 2022; 14:nu14091920. [PMID: 35565887 PMCID: PMC9103085 DOI: 10.3390/nu14091920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 12/13/2022] Open
Abstract
Hypertension (HTN) is one of the main cardiovascular risk factors and is considered a major public health problem. Numerous approaches have been developed to lower blood pressure (BP) in hypertensive patients, most of them involving pharmacological treatments. Within this context, natural bioactive compounds have emerged as a promising alternative to drugs in HTN prevention. This work reviews not only the mechanisms of BP regulation by these antihypertensive compounds, but also their efficacy depending on consumption time. Although a plethora of studies has investigated food-derived compounds, such as phenolic compounds or peptides and their impact on BP, only a few addressed the relevance of time consumption. However, it is known that BP and its main regulatory mechanisms show a 24-h oscillation. Moreover, evidence shows that phenolic compounds can interact with clock genes, which regulate the biological rhythm followed by many physiological processes. Therefore, further research might be carried out to completely elucidate the interactions along the time–nutrition–hypertension axis within the framework of chrononutrition.
Collapse
Affiliation(s)
| | | | - Enrique Calvo
- Correspondence: (E.C.); (F.I.B.); Tel.: +34-977558837 (E.C.)
| | | | | | | | | |
Collapse
|
15
|
Douma LG, Costello HM, Crislip GR, Cheng KY, Lynch IJ, Juffre A, Barral D, Masten S, Roig E, Beguiristain K, Li W, Bratanatawira P, Wingo CS, Gumz ML. Kidney-specific KO of the circadian clock protein PER1 alters renal Na + handling, aldosterone levels, and kidney/adrenal gene expression. Am J Physiol Renal Physiol 2022; 322:F449-F459. [PMID: 35129370 PMCID: PMC9169971 DOI: 10.1152/ajprenal.00385.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/20/2022] [Accepted: 01/28/2022] [Indexed: 11/22/2022] Open
Abstract
PERIOD 1 (PER1) is a circadian clock transcription factor that is regulated by aldosterone, a hormone that increases blood volume and Na+ retention to increase blood pressure. Male global Per1 knockout (KO) mice develop reduced night/day differences in Na+ excretion in response to a high-salt diet plus desoxycorticosterone pivalate treatment (HS + DOCP), a model of salt-sensitive hypertension. In addition, global Per1 KO mice exhibit higher aldosterone levels on a normal-salt diet. To determine the role of Per1 in the kidney, male kidney-specific Per1 KO (KS-Per1 KO) mice were generated using Ksp-cadherin Cre recombinase to remove exons 2-8 of Per1 in the distal nephron and collecting duct. Male KS-Per1 KO mice have increased Na+ retention but have normal diurnal differences in Na+ excretion in response to HS + DOCP. The increased Na+ retention is associated with altered expression of glucocorticoid and mineralocorticoid receptors, increased serum aldosterone, and increased medullary endothelin-1 compared with control mice. Adrenal gland gene expression analysis revealed that circadian clock and aldosterone synthesis genes have altered expression in KS-Per1 KO mice compared with control mice. These results emphasize the importance of the circadian clock not only in maintaining rhythms of physiological functions but also for adaptability in response to environmental cues, such as HS + DOCP, to maintain overall homeostasis. Given the prevalence of salt-sensitive hypertension in the general population, these findings have important implications for our understanding of how circadian clock proteins regulate homeostasis.NEW & NOTEWORTHY For the first time, we show that knockout of the circadian clock transcription factor PERIOD 1 using kidney-specific cadherin Cre results in increased renal Na+ reabsorption, increased aldosterone levels, and changes in gene expression in both the kidney and adrenal gland. Diurnal changes in renal Na+ excretion were not observed, demonstrating that the clock protein PER1 in the kidney is important for maintaining homeostasis and that this effect may be independent of time of day.
Collapse
Affiliation(s)
- Lauren G Douma
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - Hannah M Costello
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - G Ryan Crislip
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Kit-Yan Cheng
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - I Jeanette Lynch
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcolm Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Alexandria Juffre
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - Dominique Barral
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Sarah Masten
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Emilio Roig
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Kevin Beguiristain
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Wendy Li
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Phillip Bratanatawira
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Charles S Wingo
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcolm Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Michelle L Gumz
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida
| |
Collapse
|
16
|
Luan J, Yang K, Ding Y, Zhang X, Wang Y, Cui H, Zhou D, Chen L, Ma Z, Wang W, Zhang W, Liu X. Valsartan-mediated chronotherapy in spontaneously hypertensive rats via targeting clock gene expression in vascular smooth muscle cells. Arch Physiol Biochem 2022; 128:490-500. [PMID: 31794282 DOI: 10.1080/13813455.2019.1695840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE This study was to investigate the underlying mechanisms of valsartan chronotherapy in regulating blood pressure variability. METHODS RT-PCR was used to assay clock genes expression rhythm in the hypothalamus, aortic vessels, and target organs after valsartan chronotherapy. WB was used to measure Period 1 (Per1), Period 2 (Per2) protein expression in aortic vessels, as well as to measure phosphorylation of 20-kDa regulatory myosin light chain (MLC20) in VSMCs. RESULTS Specific clock genes in the hypothalamus, and Per1 and Per2 in aorta abdominalis, exhibited disordered circadian expression in vivo. Valsartan asleep time administration (VSA) restored circadian clock gene expression in a tissue- and gene-specific manner. In vitro, VSA was more efficient in blocking angiotensin II relative to VWA, which led to differential circadian rhythms of Per1 and Per2, ultimately corrected MLC20 phosphorylation. CONCLUSION VSA may be efficacious in regulating circadian clock genes rhythm, then concomitantly correct circadian blood pressure rhythms.
Collapse
Affiliation(s)
- Jiajie Luan
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Kui Yang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Yanyun Ding
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Xiaotong Zhang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Yaqin Wang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Haiju Cui
- Department of Pharmacy, XuanCheng Vocational and Technical college, XuanCheng, Anhui, P.R. China
| | - Deixi Zhou
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Lu Chen
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Zhangqing Ma
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Wusan Wang
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Wen Zhang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Xiaoyun Liu
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| |
Collapse
|
17
|
Bryant AJ, Ebrahimi E, Nguyen A, Wolff CA, Gumz ML, Liu AC, Esser KA. A wrinkle in time: circadian biology in pulmonary vascular health and disease. Am J Physiol Lung Cell Mol Physiol 2022; 322:L84-L101. [PMID: 34850650 PMCID: PMC8759967 DOI: 10.1152/ajplung.00037.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
An often overlooked element of pulmonary vascular disease is time. Cellular responses to time, which are regulated directly by the core circadian clock, have only recently been elucidated. Despite an extensive collection of data regarding the role of rhythmic contribution to disease pathogenesis (such as systemic hypertension, coronary artery, and renal disease), the roles of key circadian transcription factors in pulmonary hypertension remain understudied. This is despite a large degree of overlap in the pulmonary hypertension and circadian rhythm fields, not only including shared signaling pathways, but also cell-specific effects of the core clock that are known to result in both protective and adverse lung vessel changes. Therefore, the goal of this review is to summarize the current dialogue regarding common pathways in circadian biology, with a specific emphasis on its implications in the progression of pulmonary hypertension. In this work, we emphasize specific proteins involved in the regulation of the core molecular clock while noting the circadian cell-specific changes relevant to vascular remodeling. Finally, we apply this knowledge to the optimization of medical therapy, with a focus on sleep hygiene and the role of chronopharmacology in patients with this disease. In dissecting the unique relationship between time and cellular biology, we aim to provide valuable insight into the practical implications of considering time as a therapeutic variable. Armed with this information, physicians will be positioned to more efficiently use the full four dimensions of patient care, resulting in improved morbidity and mortality of pulmonary hypertension patients.
Collapse
Affiliation(s)
- Andrew J. Bryant
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Elnaz Ebrahimi
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Amy Nguyen
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Christopher A. Wolff
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Michelle L. Gumz
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Andrew C. Liu
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Karyn A. Esser
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| |
Collapse
|
18
|
Tsilosani A, Gao C, Zhang W. Aldosterone-Regulated Sodium Transport and Blood Pressure. Front Physiol 2022; 13:770375. [PMID: 35197862 PMCID: PMC8859437 DOI: 10.3389/fphys.2022.770375] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/06/2022] [Indexed: 11/13/2022] Open
Abstract
Aldosterone is a major mineralocorticoid steroid hormone secreted by glomerulosa cells in the adrenal cortex. It regulates a variety of physiological responses including those to oxidative stress, inflammation, fluid disruption, and abnormal blood pressure through its actions on various tissues including the kidney, heart, and the central nervous system. Aldosterone synthesis is primarily regulated by angiotensin II, K+ concentration, and adrenocorticotrophic hormone. Elevated serum aldosterone levels increase blood pressure largely by increasing Na+ re-absorption in the kidney through regulating transcription and activity of the epithelial sodium channel (ENaC). This review focuses on the signaling pathways involved in aldosterone synthesis and its effects on Na+ reabsorption through ENaC.
Collapse
Affiliation(s)
- Akaki Tsilosani
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Chao Gao
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Wenzheng Zhang
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| |
Collapse
|
19
|
Joshi H, Vastrad B, Joshi N, Vastrad C. Integrated bioinformatics analysis reveals novel key biomarkers in diabetic nephropathy. SAGE Open Med 2022; 10:20503121221137005. [PMID: 36385790 PMCID: PMC9661593 DOI: 10.1177/20503121221137005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives: The underlying molecular mechanisms of diabetic nephropathy have yet not been investigated clearly. In this investigation, we aimed to identify key genes involved in the pathogenesis and prognosis of diabetic nephropathy. Methods: We downloaded next-generation sequencing data set GSE142025 from Gene Expression Omnibus database having 28 diabetic nephropathy samples and nine normal control samples. The differentially expressed genes between diabetic nephropathy and normal control samples were analyzed. Biological function analysis of the differentially expressed genes was enriched by Gene Ontology and REACTOME pathways. Then, we established the protein–protein interaction network, modules, miRNA-differentially expressed gene regulatory network and transcription factor-differentially expressed gene regulatory network. Hub genes were validated by using receiver operating characteristic curve analysis. Results: A total of 549 differentially expressed genes were detected including 275 upregulated and 274 downregulated genes. The biological process analysis of functional enrichment showed that these differentially expressed genes were mainly enriched in cell activation, integral component of plasma membrane, lipid binding, and biological oxidations. Analyzing the protein–protein interaction network, miRNA-differentially expressed gene regulatory network and transcription factor-differentially expressed gene regulatory network, we screened hub genes MDFI, LCK, BTK, IRF4, PRKCB, EGR1, JUN, FOS, ALB, and NR4A1 by the Cytoscape software. The receiver operating characteristic curve analysis confirmed that hub genes were of diagnostic value. Conclusions: Taken above, using integrated bioinformatics analysis, we have identified key genes and pathways in diabetic nephropathy, which could improve our understanding of the cause and underlying molecular events, and these key genes and pathways might be therapeutic targets for diabetic nephropathy.
Collapse
Affiliation(s)
- Harish Joshi
- Endocrine and Diabetes Care Center, Hubbali, India
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, KLE Society’s College of Pharmacy, Gadag, India
| | - Nidhi Joshi
- Dr. D. Y. Patil Medical College, Kolhapur, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Dharwad, India
- Chanabasayya Vastrad, Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, India.
| |
Collapse
|
20
|
Abstract
Accumulating evidence suggests that the molecular circadian clock is crucial in blood pressure (BP) control. Circadian rhythms are controlled by the central clock, which resides in the suprachiasmatic nucleus of the hypothalamus and peripheral clocks throughout the body. Both light and food cues entrain these clocks but whether these cues are important for the circadian rhythm of BP is a growing area of interest. The peripheral clocks in the smooth muscle, perivascular adipose tissue, liver, adrenal gland, and kidney have been recently implicated in the regulation of BP rhythm. Dysregulation of the circadian rhythm of BP is associated with adverse cardiorenal outcomes and increased risk of cardiovascular mortality. In this review, we summarize the most recent advances in peripheral clocks as BP regulators, highlight the adverse outcomes of disrupted circadian BP rhythm in hypertension, and provide insight into potential future work in areas exploring the circadian clock in BP control and chronotherapy. A better understanding of peripheral clock function in regulating the circadian rhythm of BP will help pave the way for targeted therapeutics in the treatment of circadian BP dysregulation and hypertension.
Collapse
Affiliation(s)
- Hannah M. Costello
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32610
| | - Michelle L. Gumz
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32610
| |
Collapse
|
21
|
Fang Z, Zhu L, Jin Y, Chen Y, Chang W, Yao Y. Downregulation of Arntl mRNA Expression in Women with Hypertension: A Case-Control Study. Kidney Blood Press Res 2021; 46:741-748. [PMID: 34515147 PMCID: PMC8743905 DOI: 10.1159/000518669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/25/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Previous studies have reported that disturbance of endogenous circadian rhythms enhances the chance of hypertension and suggested that circadian clock genes could have a crucial function in the onset of the disease. This case-control study was aimed to investigate the association of the mRNA expression of aryl hydrocarbon receptor nuclear translocator like (Arntl), clock circadian regulator (Clock), and period circadian regulators 1 and 2 (Per1 and Per2) with hypertension and blood pressure levels. METHODS A total of 172 subjects were recruited in this study, including 86 hypertension and 86 nonhypertension controls. The mRNA expression levels in peripheral blood mononuclear cells were determined by real-time quantitative polymerase chain reaction. The differences in Arntl, Clock, Per1, and Per2 mRNA expression were compared between the 2 groups, and the relationship between mRNA expression and cardiometabolic risk profiles was also assessed. RESULTS We found that the mRNA expression of Arntl was downregulated in the hypertension cases compared with controls in women (1.10 [0.66, 1.71] vs. 1.30 [0.99, 2.06], p = 0.031). There was a significant negative correlation between the Arntl mRNA expression and SBP (r = -0.301, p = 0.004) and DBP (r = -0.222, p = 0.034) in women. In men, a negative correlation between the Per1 mRNA expression and SBP (r = -0.247, p = 0.026) was found. CONCLUSIONS The Arntl mRNA expression may play an important role in progression of hypertension in women.
Collapse
Affiliation(s)
- Zhengmei Fang
- Department of Epidemiology, School of Public Health, and Institute of Chronic Disease Prevention and Control, Wannan Medical College, Wuhu, China
| | - Lijun Zhu
- Department of Epidemiology, School of Public Health, and Institute of Chronic Disease Prevention and Control, Wannan Medical College, Wuhu, China
| | - Yuelong Jin
- Department of Epidemiology, School of Public Health, and Institute of Chronic Disease Prevention and Control, Wannan Medical College, Wuhu, China
| | - Yan Chen
- Department of Epidemiology, School of Public Health, and Institute of Chronic Disease Prevention and Control, Wannan Medical College, Wuhu, China
| | - Weiwei Chang
- Department of Epidemiology, School of Public Health, and Institute of Chronic Disease Prevention and Control, Wannan Medical College, Wuhu, China
| | - Yingshui Yao
- Department of Epidemiology, School of Public Health, and Institute of Chronic Disease Prevention and Control, Wannan Medical College, Wuhu, China
- Anhui College of Traditional Chinese Medicine, Wuhu, China
| |
Collapse
|
22
|
Meléndez-Fernández OH, Walton JC, DeVries AC, Nelson RJ. Clocks, Rhythms, Sex, and Hearts: How Disrupted Circadian Rhythms, Time-of-Day, and Sex Influence Cardiovascular Health. Biomolecules 2021; 11:883. [PMID: 34198706 PMCID: PMC8232105 DOI: 10.3390/biom11060883] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases are the top cause of mortality in the United States, and ischemic heart disease accounts for 16% of all deaths around the world. Modifiable risk factors such as diet and exercise have often been primary targets in addressing these conditions. However, mounting evidence suggests that environmental factors that disrupt physiological rhythms might contribute to the development of these diseases, as well as contribute to increasing other risk factors that are typically associated with cardiovascular disease. Exposure to light at night, transmeridian travel, and social jetlag disrupt endogenous circadian rhythms, which, in turn, alter carefully orchestrated bodily functioning, and elevate the risk of disease and injury. Research into how disrupted circadian rhythms affect physiology and behavior has begun to reveal the intricacies of how seemingly innocuous environmental and social factors have dramatic consequences on mammalian physiology and behavior. Despite the new focus on the importance of circadian rhythms, and how disrupted circadian rhythms contribute to cardiovascular diseases, many questions in this field remain unanswered. Further, neither time-of-day nor sex as a biological variable have been consistently and thoroughly taken into account in previous studies of circadian rhythm disruption and cardiovascular disease. In this review, we will first discuss biological rhythms and the master temporal regulator that controls these rhythms, focusing on the cardiovascular system, its rhythms, and the pathology associated with its disruption, while emphasizing the importance of the time-of-day as a variable that directly affects outcomes in controlled studies, and how temporal data will inform clinical practice and influence personalized medicine. Finally, we will discuss evidence supporting the existence of sex differences in cardiovascular function and outcomes following an injury, and highlight the need for consistent inclusion of both sexes in studies that aim to understand cardiovascular function and improve cardiovascular health.
Collapse
Affiliation(s)
- O. Hecmarie Meléndez-Fernández
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (J.C.W.); (R.J.N.)
| | - James C. Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (J.C.W.); (R.J.N.)
| | - A. Courtney DeVries
- Department of Medicine, Division of Oncology/Hematology, West Virginia University, Morgantown, WV 26505, USA;
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26505, USA
| | - Randy J. Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (J.C.W.); (R.J.N.)
| |
Collapse
|
23
|
Zhang J, Sun R, Jiang T, Yang G, Chen L. Circadian Blood Pressure Rhythm in Cardiovascular and Renal Health and Disease. Biomolecules 2021; 11:biom11060868. [PMID: 34207942 PMCID: PMC8230716 DOI: 10.3390/biom11060868] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 12/21/2022] Open
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.
Collapse
Affiliation(s)
- Jiayang Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China; (J.Z.); (R.S.); (T.J.)
| | - Ruoyu Sun
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China; (J.Z.); (R.S.); (T.J.)
| | - Tingting Jiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China; (J.Z.); (R.S.); (T.J.)
| | - Guangrui Yang
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China;
| | - Lihong Chen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China; (J.Z.); (R.S.); (T.J.)
- Correspondence: ; Tel.: +86-411-86118984
| |
Collapse
|
24
|
Škrlec I, Talapko J, Juzbašić M, Steiner R. Sex Differences in Circadian Clock Genes and Myocardial Infarction Susceptibility. J Cardiovasc Dev Dis 2021; 8:53. [PMID: 34066863 PMCID: PMC8151899 DOI: 10.3390/jcdd8050053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/23/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
The growing body of evidence shows a significant difference in the circadian rhythm of cardiovascular disease based on biological sex. The incidence of cardiovascular disease varies between women and men. Additionally, biological sex is vital for the timely application of therapy-chronotherapy, which benefits both sexes. This study aimed to examine the potential difference of single nucleotide polymorphisms (SNPs) of the circadian rhythm genes ARNTL, CLOCK, CRY2 and PER2 in women and men with myocardial infarction. A cross-sectional study was conducted, including 200 patients with myocardial infarction. Altogether, ten single nucleotide polymorphisms in the ARNTL, CLOCK, CRY2 and PER2 genes were analyzed. The Chi-square test yielded statistically significant differences in CLOCK gene rs11932595 polymorphism in a recessive genotype model between women and men with a p-value of 0.03 and an odds ratio 2.66, and a corresponding 95% confidence interval of 1.07 to 6.66. Other analyzed polymorphisms of the circadian rhythm genes ARNTL, CRY2, and PER2 did not significantly differ between the sexes. According to the study's current results, the CLOCK gene's genetic variability might affect myocardial infarction concerning biological sex.
Collapse
Affiliation(s)
- Ivana Škrlec
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (J.T.); (M.J.)
| | - Jasminka Talapko
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (J.T.); (M.J.)
| | - Martina Juzbašić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (J.T.); (M.J.)
| | - Robert Steiner
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Josipa Huttlera 4, HR-31000 Osijek, Croatia;
- Clinical Department of Cardiovascular Diseases and Intensive Care, Clinic for Internal Medicine, University Hospital Osijek, Josipa Huttlera 4, HR-31000 Osijek, Croatia
| |
Collapse
|
25
|
Lin Y, Tsai M, Hsieh I, Wen M, Wang C. Deficiency of circadian gene cryptochromes in bone marrow‐derived cells protects against atherosclerosis in
LDLR
−/−
mice. FASEB J 2021; 35:e21309. [DOI: 10.1096/fj.202001818rrr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Yu‐Sheng Lin
- Healthcare Center Chang Gung Memorial Hospital Chang Gung University College of Medicine Taoyuan City Taiwan
- Department of Cardiology Chang Gung Memorial Hospital Chang Gung University College of Medicine Taoyuan City Taiwan
| | - Ming‐Lung Tsai
- Department of Cardiology Chang Gung Memorial Hospital Chang Gung University College of Medicine Taoyuan City Taiwan
| | - I‐Chang Hsieh
- Department of Cardiology Chang Gung Memorial Hospital Chang Gung University College of Medicine Taoyuan City Taiwan
| | - Ming‐Shien Wen
- Department of Cardiology Chang Gung Memorial Hospital Chang Gung University College of Medicine Taoyuan City Taiwan
| | - Chao‐Yung Wang
- Department of Cardiology Chang Gung Memorial Hospital Chang Gung University College of Medicine Taoyuan City Taiwan
- Institute of Cellular and System Medicine National Health Research Institutes Zhunan Taiwan
- Department of Medical Science National Tsing Hua University Hsinchu Taiwan
| |
Collapse
|
26
|
Douma LG, Barral D, Gumz ML. Interplay of the Circadian Clock and Endothelin System. Physiology (Bethesda) 2021; 36:35-43. [PMID: 33325818 DOI: 10.1152/physiol.00021.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The peptide hormone endothelin-1 and its receptors are linked to several disease states. Pharmacological inhibition of this pathway has proven beneficial in pulmonary hypertension, yet its potential in other disease states remains to be realized. This review considers an often understudied aspect of endothelin biology, circadian rhythm regulation and how understanding the intersection between endothelin signaling and the circadian clock may be leveraged to realize the potential of endothelin-based therapeutics.
Collapse
Affiliation(s)
- Lauren G Douma
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida.,Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - Dominique Barral
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
| | - Michelle L Gumz
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida.,Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida.,Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida.,Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, Florida
| |
Collapse
|
27
|
Ivy JR, Bailey MA. Nondipping Blood Pressure: Predictive or Reactive Failure of Renal Sodium Handling? Physiology (Bethesda) 2021; 36:21-34. [PMID: 33325814 DOI: 10.1152/physiol.00024.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Blood pressure follows a daily rhythm, dipping during nocturnal sleep in humans. Attenuation of this dip (nondipping) is associated with increased risk of cardiovascular disease. Renal control of sodium homeostasis is essential for long-term blood pressure control. Sodium reabsorption and excretion have rhythms that rely on predictive/circadian as well as reactive adaptations. We explore how these rhythms might contribute to blood pressure rhythm in health and disease.
Collapse
Affiliation(s)
- Jessica R Ivy
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Matthew A Bailey
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
28
|
Kanki M, Young MJ. Corticosteroids and circadian rhythms in the cardiovascular system. Curr Opin Pharmacol 2020; 57:21-27. [PMID: 33207294 DOI: 10.1016/j.coph.2020.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/19/2022]
Abstract
The mineralocorticoid receptor (MR) plays a central role in cardiac physiological function and disease and is thus an attractive therapeutic target for patients with heart failure. However, the incidence of significant side effects from mineralocorticoid receptor antagonist (MRA) treatment has led to investigation of new mechanisms that may enhance MR targeted therapies. Recent studies have identified the circadian clock as a novel, reciprocal interacting partner of the MR in the heart. While the closely related glucocorticoid receptor (GR) and its ligand, cortisol (corticosterone in rodents), are established regulators of the circadian clock, new data suggest that the MR can also regulate circadian clock gene expression and timing. This review will discuss the role of the MR and its ligands in the regulation of the circadian clock in the heart and the implications of dysregulation of these systems for cardiac disease progression, and for MR activation.
Collapse
Affiliation(s)
- Monica Kanki
- Cardiovascular Endocrinology Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia; Cardiovascular Endocrinology Laboratory, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular & Translational Science, Monash University, Clayton, VIC, Australia
| | - Morag J Young
- Cardiovascular Endocrinology Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia; Cardiovascular Endocrinology Laboratory, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular & Translational Science, Monash University, Clayton, VIC, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
29
|
Pearson JA, Wong FS, Wen L. Crosstalk between circadian rhythms and the microbiota. Immunology 2020; 161:278-290. [PMID: 33090484 PMCID: PMC7692254 DOI: 10.1111/imm.13278] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 09/20/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
Circadian rhythms influence daily molecular oscillations in gene/protein expression and aspects of biology and physiology, including behaviour, body temperature and sleep–wake cycles. These circadian rhythms have been associated with a number of metabolic, immune and microbial changes that correlate with health and susceptibility to disease, including infection. While light is the main inducer of circadian rhythms, other factors, including the microbiota, can have important effects on peripheral rhythms. The microbiota have been of significant interest to many investigators over the past decade, with the development of molecular techniques to identify large numbers of species and their function. These studies have shown microbial associations with disease susceptibility, and some of these have demonstrated that alterations in microbiota cause disease. Microbial circadian oscillations impact host metabolism and immunity directly and indirectly. Interestingly, microbial oscillations also regulate host circadian rhythms, and the host circadian rhythms in turn modulate microbial composition. Thus, it is of considerable interest and importance to understand the crosstalk between circadian rhythms and microbiota and especially the microbial influences on the host. In this review, we aim to discuss the role of circadian microbial oscillations and how they influence host immunity. In addition, we discuss how host circadian rhythms can also modulate microbial rhythms. We also discuss potential connections between microbes and circadian rhythms and how these may be used therapeutically to maximize clinical success.
Collapse
Affiliation(s)
- James Alexander Pearson
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK.,Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Florence Susan Wong
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Li Wen
- Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
30
|
Douma LG, Crislip GR, Cheng KY, Barral D, Masten S, Holzworth M, Roig E, Glasford K, Beguiristain K, Li W, Bratanatawira P, Lynch IJ, Cain BD, Wingo CS, Gumz ML. Knockout of the circadian clock protein PER1 results in sex-dependent alterations of ET-1 production in mice in response to a high-salt diet plus mineralocorticoid treatment. Can J Physiol Pharmacol 2020; 98:579-586. [PMID: 32437627 DOI: 10.1139/cjpp-2019-0688] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Previously, we showed that global knockout (KO) of the circadian clock transcription factor PER1 in male, but not female, mice fed a high-salt diet plus mineralocorticoid treatment (HS/DOCP) resulted in nondipping hypertension and decreased night/day ratio of sodium (Na) excretion. Additionally, we have shown that the endothelin-1 (ET-1) gene is targeted by both PER1 and aldosterone. We hypothesized that ET-1 would exhibit a sex-specific response to HS/DOCP treatment in PER1 KO. Here we show that male, but not female, global PER1 KO mice exhibit a decreased night/day ratio of urinary ET-1. Gene expression analysis revealed significant genotype differences in ET-1 and endothelin A receptor (ETA) expression in male, but not female, mice in response to HS/DOCP. Additionally, both wild-type and global PER1 KO male mice significantly increase endothelin B receptor (ETB) expression in response to HS/DOCP, but female mice do not. Finally, siRNA-mediated knockdown of PER1 in mouse cortical collecting duct cells (mpkCCDc14) resulted in increased ET-1 mRNA expression and peptide secretion in response to aldosterone treatment. These data suggest that PER1 is a negative regulator of ET-1 expression in response to HS/DOCP, revealing a novel mechanism for the regulation of renal Na handling in response to HS/DOCP treatment.
Collapse
Affiliation(s)
- Lauren G Douma
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
| | - G Ryan Crislip
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
| | - Kit-Yan Cheng
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
| | - Dominique Barral
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
| | - Sarah Masten
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
| | - Meaghan Holzworth
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
| | - Emilio Roig
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
| | - Krystal Glasford
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
| | - Kevin Beguiristain
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
| | - Wendy Li
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
| | - Phillip Bratanatawira
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
| | - I Jeanette Lynch
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA.,North Florida/South Georgia Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL 32611, USA
| | - Brian D Cain
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
| | - Charles S Wingo
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA.,North Florida/South Georgia Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL 32611, USA
| | - Michelle L Gumz
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA.,Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA.,North Florida/South Georgia Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL 32611, USA
| |
Collapse
|
31
|
de Souza AMA, Ji H, Wu X, Sandberg K, West CA. Persistent Renin-Angiotensin System Sensitization Months After Body Weight Recovery From Severe Food Restriction in Female Fischer Rats. J Am Heart Assoc 2020; 9:e017246. [PMID: 32674648 PMCID: PMC7660733 DOI: 10.1161/jaha.120.017246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Prior exposure to periods of severe food restriction (sFR) is associated with increased risk of developing hypertension and cardiovascular disease later in life. Methods and Results To investigate the mechanism of these long-term adverse effects of sFR, 4-month-old female Fischer rats were divided in 2 groups and maintained on a normal diet ad libitum (control) or on an sFR diet with 60% reduction in daily food intake for 2 weeks that resulted in a 15% reduction in body weight. After the 2-week sFR period ended, both groups received normal chow ad libitum for 3 months. Within 2 weeks after refeeding was initiated in the sFR group, body weight was restored to control levels; however, plasma angiotensinogen (1.3-fold; P<0.05), Ang-[1-8] (2.0-fold; P<0.05), and angiotensin-converting enzyme activity (1.1-fold; P<0.01) were all elevated 3 months after refeeding. Angiotensin type 1 receptor activity was also increased as evidenced by augmented pressor responses to angiotensin-[1-8] (P<0.01) and depressor responses to the angiotensin type 1 receptor antagonist, losartan (P<0.01) in the sFR group. Conclusions These results indicate that sensitization of the renin-angiotensin system persisted months after the sFR period ended. These findings may have implications for women who voluntarily or involuntarily experience an extended period of sFR and thus may be at increased risk of developing cardiovascular disease through sensitization of the renin-angiotensin system even though their body weight, mean arterial pressure, and heart rate appear normal.
Collapse
Affiliation(s)
| | - Hong Ji
- Department of Medicine Georgetown University Washington DC
| | - Xie Wu
- Department of Medicine Georgetown University Washington DC
| | | | - Crystal A West
- Department of Medicine Georgetown University Washington DC
| |
Collapse
|
32
|
Johnston JG, Speed JS, Becker BK, Kasztan M, Soliman RH, Rhoads MK, Tao B, Jin C, Geurts AM, Hyndman KA, Pollock JS, Pollock DM. Diurnal Control of Blood Pressure Is Uncoupled From Sodium Excretion. Hypertension 2020; 75:1624-1634. [PMID: 32306766 PMCID: PMC7228023 DOI: 10.1161/hypertensionaha.119.13908] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The diurnal rhythms of sodium handling and blood pressure are thought to be regulated by clock genes, such as Bmal1. However, little is known about the regulation of these factors by Bmal1, especially in rats. Using a novel whole-body Bmal1 knockout rat model (Bmal1-/-), we hypothesized that time of day regulation of sodium excretion is dependent on Bmal1. Using telemetry to continuously record mean arterial pressure, we observed that male and female Bmal1-/- rats had significantly reduced mean arterial pressure over the course of 24 hours compared with littermate controls. The circadian mean arterial pressure pattern remained intact in both sexes of Bmal1-/- rats, which is in contrast to the Bmal1-/- mouse model. Male Bmal1-/- rats had no significant difference in baseline sodium excretion between 12-hour active and inactive periods, indicating a lack of diurnal control independent of maintained mean arterial pressure rhythms. Female Bmal1-/- rats, however, had significantly greater sodium excretion during the active versus inactive period similar to controls. Thus, we observed a clear dissociation between circadian blood pressure and control of sodium excretion that is sex dependent. These findings are consistent with a more robust ability of females to maintain control of sodium excretion, and furthermore, demonstrate a novel role for Bmal1 in control of diurnal blood pressure independent of sodium excretion.
Collapse
Affiliation(s)
- Jermaine G. Johnston
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Joshua S. Speed
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216
| | - Bryan K. Becker
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Malgorzata Kasztan
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Reham H. Soliman
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Megan K. Rhoads
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Binli Tao
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Chunhua Jin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Aron M. Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Kelly A. Hyndman
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jennifer S. Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - David M. Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
33
|
Crislip GR, Douma LG, Masten SH, Cheng KY, Lynch IJ, Johnston JG, Barral D, Glasford KB, Holzworth MR, Verlander JW, Wingo CS, Gumz ML. Differences in renal BMAL1 contribution to Na + homeostasis and blood pressure control in male and female mice. Am J Physiol Renal Physiol 2020; 318:F1463-F1477. [PMID: 32338037 PMCID: PMC7311713 DOI: 10.1152/ajprenal.00014.2020] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/07/2020] [Accepted: 04/20/2020] [Indexed: 12/27/2022] Open
Abstract
The renal circadian clock has a major influence on the function of the kidney. Aryl hydrocarbon receptor nuclear translocator-like protein 1 [ARNTL; also known as brain and muscle ARNT-like 1 (BMAL1)] is a core clock protein and transcription factor that regulates the expression of nearly half of all genes. Using male and female kidney-specific cadherin BMAL1 knockout (KS-BMAL1 KO) mice, we examined the role of renal distal segment BMAL1 in blood pressure control and solute handling. We confirmed that this mouse model does not express BMAL1 in thick ascending limb, distal convoluted tubule, and collecting duct cells, which are the final locations for solute and fluid regulation. Male KS-BMAL1 KO mice displayed a substantially lower basal systolic blood pressure compared with littermate control mice, yet their circadian rhythm in pressure remained unchanged [male control mice: 127 ± 0.7 mmHg (n = 4) vs. male KS-BMAL KO mice: 119 ± 2.3 mmHg (n = 5), P < 0.05]. Female mice, however, did not display a genotype difference in basal systolic blood pressure [female control mice: 120 ± 1.6 mmHg (n = 5) vs. female KS-BMAL1 KO mice: 119 ± 1.5 mmHg (n = 7), P = 0.4]. In addition, male KS-BMAL1 KO mice had less Na+ retention compared with control mice in response to a K+-restricted diet (15% less following 5 days of treatment). However, there was no genotype difference in Na+ handling after a K+-restricted diet in female mice. Furthermore, there was evidence indicating a sex-specific response to K+ restriction where female mice reabsorbed less Na+ in response to this dietary challenge compared with male mice. We propose that BMAL1 in the distal nephron and collecting duct contributes to blood pressure regulation and Na+ handling in a sex-specific manner.
Collapse
Affiliation(s)
- G Ryan Crislip
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
| | - Lauren G Douma
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - Sarah H Masten
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
| | - Kit-Yan Cheng
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
| | - I Jeanette Lynch
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
- North Florida/South Georgia Veterans Health System, Gainesville, Florida
| | - Jermaine G Johnston
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
| | - Dominique Barral
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
| | - Krystal B Glasford
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
| | - Meaghan R Holzworth
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
| | - Jill W Verlander
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
| | - Charles S Wingo
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
- North Florida/South Georgia Veterans Health System, Gainesville, Florida
| | - Michelle L Gumz
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| |
Collapse
|
34
|
Chakraborty S, Mandal J, Cheng X, Galla S, Hindupur A, Saha P, Yeoh BS, Mell B, Yeo JY, Vijay-Kumar M, Yang T, Joe B. Diurnal Timing Dependent Alterations in Gut Microbial Composition Are Synchronously Linked to Salt-Sensitive Hypertension and Renal Damage. Hypertension 2020; 76:59-72. [PMID: 32450738 DOI: 10.1161/hypertensionaha.120.14830] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alterations of diurnal rhythms of blood pressure (BP) and reshaping of gut microbiota are both independently associated with hypertension. However, the relationships between biorhythms of BP and gut microbial composition are unknown. We hypothesized that diurnal timing-associated alterations of microbial compositions are synchronous with diurnal rhythmicity, dip in BP, and renal function. To test this hypothesis, Dahl salt-sensitive (S) rats on low- and high-salt diets were examined for time of day effects on gut microbiota, BP, and indicators of renal damage. Major shifts in night and day patterns of specific groups of microbiota were observed between the dark (active) and light (rest) phases, which correlated with diurnal rhythmicity of BP. The diurnal abundance of Firmicutes, Bacteroidetes, and Actinobacteria were independently associated with BP. Discrete bacterial taxa were observed to correlate independently or interactively with one or more of the following 3 factors: (1) BP rhythm, (2) dietary salt, and (3) dip in BP. Phylogenetic Investigation of Communities revealed diurnal timing effects on microbial pathways, characterized by upregulated biosynthetic processes during the active phase of host, and upregulated degradation pathways of metabolites in the resting phase. Additional metagenomics functional pathways with rhythm variations were noted for aromatic amino acid metabolism and taurine metabolism. These diurnal timing dependent changes in microbiota, their functional pathways, and BP dip were associated with concerted effects of the levels of renal lipocalin 2 and kidney injury molecule-1 expression. These data provide evidence for a firm and concerted diurnal timing effects of BP, renal damage, and select microbial communities.
Collapse
Affiliation(s)
- Saroj Chakraborty
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| | - Juthika Mandal
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| | - Xi Cheng
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| | - Sarah Galla
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| | - Anay Hindupur
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| | - Piu Saha
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| | - Beng San Yeoh
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| | - Blair Mell
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| | - Ji-Youn Yeo
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| | - Matam Vijay-Kumar
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| | - Tao Yang
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| | - Bina Joe
- From the Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH; and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH (S.J., J.M., X.C., S.G., A.H., P.S., B.S.Y., B.M., J.-Y.Y., M.V-K., T.Y., B.J.)
| |
Collapse
|
35
|
Soares AC, Fonseca DA. Cardiovascular diseases: a therapeutic perspective around the clock. Drug Discov Today 2020; 25:1086-1098. [PMID: 32320853 DOI: 10.1016/j.drudis.2020.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 01/21/2023]
Abstract
Biological rhythms are a ubiquitous feature of life. Most bodily functions, including physiological, biochemical, and behavioral processes, are coupled by the circadian rhythm. In the cardiovascular system, circadian fluctuations regulate several functions, namely heart rate, blood pressure, cardiac contractility, and metabolism. In fact, current lifestyles impose external timing constraints that clash with our internal circadian physiology, often increasing the risk of cardiovascular disease (CVD). Still, the mechanisms of dysregulation are not fully understood because this is a growing area of research. In this review, we explore the modulatory role of the circadian rhythms on cardiovascular function and disease as well as the role of chronotherapy in the context of CVD and how such an approach could improve existing therapies and assist in the development of new ones.
Collapse
Affiliation(s)
| | - Diogo A Fonseca
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; CIBB Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal.
| |
Collapse
|
36
|
Zhang D, Jin C, Obi IE, Rhoads MK, Soliman RH, Sedaka RS, Allan JM, Tao B, Speed JS, Pollock JS, Pollock DM. Loss of circadian gene Bmal1 in the collecting duct lowers blood pressure in male, but not female, mice. Am J Physiol Renal Physiol 2020; 318:F710-F719. [PMID: 31904281 PMCID: PMC7099501 DOI: 10.1152/ajprenal.00364.2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 12/11/2019] [Accepted: 01/03/2020] [Indexed: 01/19/2023] Open
Abstract
Kidney function follows a 24-h rhythm subject to regulation by circadian genes including the transcription factor Bmal1. A high-salt diet induces a phase shift in Bmal1 expression in the renal inner medulla that is dependent on endothelin type B (ETB) receptors. Furthermore, ETB receptor-mediated natriuresis is sex dependent. Therefore, experiments tested the hypothesis that collecting duct Bmal1 regulates blood pressure in a sex-dependent manner. We generated a mouse model that lacks Bmal1 expression in the collecting duct, where ETB receptor abundance is highest. Male, but not female, collecting duct Bmal1 knockout (CDBmal1KO) mice had significantly lower 24-h mean arterial pressure (MAP) than flox controls (105 ± 2 vs. 112 ± 3 mmHg for male mice and 106 ± 1 vs. 108 ± 1 mmHg for female mice, by telemetry). After 6 days on a high-salt (4% NaCl) diet, MAP remained significantly lower in male CDBmal1KO mice than in male flox control mice (107 ± 2 vs. 113 ± 1 mmHg), with no significant differences between genotypes in female mice (108 ± 2 vs. 109 ± 1 mmHg). ETB receptor blockade for another 6 days increased MAP similarly in both male and female CDBmal1KO and flox control mice. However, MAP remained lower in male CDBmal1KO mice than in male flox control mice (124 ± 2 vs. 130 ± 2 mmHg). No significant differences were observed between female CDBmal1KO and flox mice during ETB blockade (130 ± 2 vs. 127 ± 2 mmHg). There were no significant genotype differences in amplitude or phase of MAP in either sex. These data suggest that collecting duct Bmal1 has no role in circadian MAP but plays an important role in overall blood pressure in male, but not female, mice.
Collapse
Affiliation(s)
- Dingguo Zhang
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Chunhua Jin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ijeoma E Obi
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Megan K Rhoads
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Reham H Soliman
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Randee S Sedaka
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - J Miller Allan
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Binli Tao
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Joshua S Speed
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jennifer S Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David M Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
37
|
Zhang D, Pollock DM. Diurnal Regulation of Renal Electrolyte Excretion: The Role of Paracrine Factors. Annu Rev Physiol 2019; 82:343-363. [PMID: 31635525 DOI: 10.1146/annurev-physiol-021119-034446] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many physiological processes, including most kidney-related functions, follow specific rhythms tied to a 24-h cycle. This is largely because circadian genes operate in virtually every cell type in the body. In addition, many noncanonical genes have intrinsic circadian rhythms, especially within the liver and kidney. This new level of complexity applies to the control of renal electrolyte excretion. Furthermore, there is growing evidence that paracrine and autocrine factors, especially the endothelin system, are regulated by clock genes. We have known for decades that excretion of electrolytes is dependent on time of day, which could play an important role in fluid volume balance and blood pressure control. Here, we review what is known about the interplay between paracrine and circadian control of electrolyte excretion. The hope is that recognition of paracrine and circadian factors can be considered more deeply in the future when integrating with well-established neuroendocrine control of excretion.
Collapse
Affiliation(s)
- Dingguo Zhang
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35233, USA; ,
| | - David M Pollock
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35233, USA; ,
| |
Collapse
|
38
|
Zhao Q, Sun H, Yin L, Wang L. miR‑126a‑5p‑Dbp and miR‑31a‑Crot/Mrpl4 interaction pairs crucial for the development of hypertension and stroke. Mol Med Rep 2019; 20:4151-4167. [PMID: 31545431 PMCID: PMC6797943 DOI: 10.3892/mmr.2019.10679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/19/2019] [Indexed: 01/09/2023] Open
Abstract
The present study aimed to integrate the mRNA and microRNA (miRNA) expression profiles of spontaneously hypertensive rats (SHR rats) and stroke-prone spontaneously hypertensive rats (SHRSP rats) to screen for potential therapeutic targets for hypertension and stroke. The datasets GSE41452, GSE31457, GSE41453 and GSE53363 were collected from the Gene Expression Omnibus (GEO) database to screen differentially expressed genes (DEGs). The GSE53361 dataset was obtained to analyze differentially expressed miRNAs (DEMs). The DEGs and DEMs were identified between SHR (or SHRSP) rats and normotensive Wistar-Kyoto (WKY) rats using the Linear Models for Microarray (limma) data method. Venn diagrams were used to show the SHR-specific, SHRSP-specific and SHR-SHRSP shared DEGs and DEMs, and these were utilized to construct the protein-protein interaction (PPI) and miRNA-mRNA regulatory networks. The Database for Annotation, Visualization and Integrated Discovery (DAVID) was used to explore the function of the genes. Subsequently, the connectivity Map (CMAP) database was searched to identify small-molecule drugs. Comparisons between the GSE41452-GSE31457-GSE41453 merged and GSE53363 datasets identified 2 SHR-specific, 8 SHRSP-specific and 15 SHR-SHRSP shared DEGs. Function enrichment analysis showed that SHRSP-specific D-box binding PAR bZIP transcription factor (Dbp) was associated with circadian rhythm, and SHR-SHRSP shared carnitine O-octanoyltransferase (Crot) was involved in fatty acid metabolic processes or the inflammatory response via interacting with epoxide hydrolase 2 (EPHX2). SHR-SHRSP shared mitochondrial ribosomal protein L4 (Mrpl4) may exert roles by interacting with the threonine-tRNA ligase, TARS2. The miRNA regulatory network predicted that upregulated Dbp could be regulated by rno-miR-126a-5p, whereas downregulated Crot and Mrpl4 could be modulated by rno-miR-31a. The CMAP database predicted that small-molecule drugs, including botulin, Gly-His-Lys, and podophyllotoxin, may possess therapeutic potential. In conclusion, the present study has identified Dbp, Crot and Mrpl4 as potential targets for the treatment of hypertension and stroke. Furthermore, the expression of these genes may be reversed by the above miRNAs or drugs.
Collapse
Affiliation(s)
- Qini Zhao
- Department of Cardiology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Huan Sun
- Department of Cardiology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Liquan Yin
- Department of Rehabilitation Medicine, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Libo Wang
- Department of Neurology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
39
|
Alli A, Yu L, Holzworth M, Richards J, Cheng KY, Lynch IJ, Wingo CS, Gumz ML. Direct and indirect inhibition of the circadian clock protein Per1: effects on ENaC and blood pressure. Am J Physiol Renal Physiol 2019; 316:F807-F813. [PMID: 30759025 DOI: 10.1152/ajprenal.00408.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Circadian rhythms govern physiological functions and are important for overall health. The molecular circadian clock comprises several transcription factors that mediate circadian control of physiological function, in part, by regulating gene expression in a tissue-specific manner. These connections are well established, but the underlying mechanisms are incompletely understood. The overall goal of this study was to examine the connection among the circadian clock protein Period 1 (Per1), epithelial Na+ channel (ENaC), and blood pressure (BP) using a multipronged approach. Using global Per1 knockout mice on a 129/sv background in combination with a high-salt diet plus mineralocorticoid treatment, we demonstrated that loss of Per1 in this setting is associated with protection from hypertension. Next, we used the ENaC inhibitor benzamil to demonstrate a role for ENaC in BP regulation and urinary Na+ excretion in 129/sv mice. We targeted Per1 indirectly using pharmacological inhibition of Per1 nuclear entry in vivo to demonstrate altered expression of known Per1 target genes as well as a BP-lowering effect in 129/sv mice. Finally, we directly inhibited Per1 via genetic knockdown in amphibian distal nephron cells to demonstrate, for the first time, that reduced Per1 expression is associated with decreased ENaC activity at the single channel level.
Collapse
Affiliation(s)
- Abdel Alli
- Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida.,Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida , Gainesville, Florida
| | - Ling Yu
- Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida.,College of Resources and Environmental Sciences, Nanjing Agricultural University , Nanjing , China
| | - Meaghan Holzworth
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida , Gainesville, Florida
| | - Jacob Richards
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida , Gainesville, Florida
| | - Kit-Yan Cheng
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida , Gainesville, Florida.,North Florida/South Georgia Veterans Affairs Medical Center , Gainesville, Florida
| | - I Jeanette Lynch
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida , Gainesville, Florida.,North Florida/South Georgia Veterans Affairs Medical Center , Gainesville, Florida
| | - Charles S Wingo
- Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida.,Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida , Gainesville, Florida.,North Florida/South Georgia Veterans Affairs Medical Center , Gainesville, Florida
| | - Michelle L Gumz
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida , Gainesville, Florida.,North Florida/South Georgia Veterans Affairs Medical Center , Gainesville, Florida.,Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida
| |
Collapse
|