1
|
Myung J, Vitet H, Truong VH, Ananthasubramaniam B. The role of the multiplicity of circadian clocks in mammalian systems. Sleep Med 2025; 131:106518. [PMID: 40222295 DOI: 10.1016/j.sleep.2025.106518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/22/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
Circadian clocks regulate rhythmic biological processes in nearly every tissue, aligning physiology and behavior with the 24-h light-dark cycle. While the central circadian clock in the suprachiasmatic nucleus (SCN) has been extensively studied, emerging evidence indicates that virtually every cell in the body possesses its own locally autonomous circadian clock. This raises a fundamental question: why do multicellular organisms utilize multiple circadian clocks instead of a single master clock broadcasting time cues? Here, we examine how distributed local clocks differ from phase-resettable cycles and ensure robust temporal scheduling of physiological processes. We discuss how internal entrainment among local clocks governs self-sustained, yet flexible, circadian organization of tissue-specific responses to environmental changes. We also examine how the organization of clocks contributes to seasonal homeostasis, and the implications for disease when coordination among these clocks is disrupted.
Collapse
Affiliation(s)
- Jihwan Myung
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Hélène Vitet
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan
| | - Vuong Hung Truong
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan
| | | |
Collapse
|
2
|
Preston R, Chrisp R, Dudek M, Morais MRPT, Tian P, Williams E, Naylor RW, Davenport B, Pathiranage DRJ, Benson E, Spiller DG, Bagnall J, Zeef L, Lawless C, Baker SM, Meng QJ, Lennon R. The glomerular circadian clock temporally regulates basement membrane dynamics and the podocyte glucocorticoid response. Kidney Int 2025; 107:99-115. [PMID: 39515644 DOI: 10.1016/j.kint.2024.10.016] [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: 12/01/2023] [Revised: 09/19/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
Kidney physiology shows diurnal variation, and a disrupted circadian rhythm is associated with kidney disease. However, it remains largely unknown whether glomeruli, the filtering units in the kidney, are under circadian control. Here, we investigated core circadian clock components in glomeruli, together with their rhythmic targets and modes of regulation. With clock gene reporter mice, cell-autonomous glomerular clocks which likely govern rhythmic fluctuations in glomerular physiology were identified. Using circadian time-series transcriptomic profiling, the first circadian glomerular transcriptome with 375 rhythmic transcripts, enriched for extracellular matrix and glucocorticoid receptor signaling ontologies, were identified. Subsets of rhythmic matrix-related genes required for basement membrane assembly and turnover, and circadian variation in matrix ultrastructure, coinciding with peak abundance of rhythmic basement membrane proteins, were uncovered. This provided multiomic evidence for interactions between glomerular matrix and intracellular time-keeping mechanisms. Furthermore, glucocorticoids, which are frequently used to treat glomerular disease, reset the podocyte clock and induce rhythmic expression of potential glomerular disease genes associated with nephrotic syndrome that included Nphs1 (nephrin) and Nphs2 (podocin). Disruption of the clock with pharmacological inhibition altered the expression of these disease genes, indicating an interplay between clock gene expression and key genes required for podocyte health. Thus, our results provide a strong basis for future investigations of the functional implications and therapeutic potential of chronotherapy in glomerular health and disease.
Collapse
Affiliation(s)
- Rebecca Preston
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Ruby Chrisp
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Michal Dudek
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Mychel R P T Morais
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Pinyuan Tian
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Emily Williams
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Richard W Naylor
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Bernard Davenport
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Dharshika R J Pathiranage
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Emma Benson
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - David G Spiller
- Bioimaging Core Facility, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - James Bagnall
- Bioimaging Core Facility, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - Leo Zeef
- Bioinformatics Core Facility, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - Craig Lawless
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Syed Murtuza Baker
- Bioinformatics Core Facility, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - Qing-Jun Meng
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Department of Pediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
3
|
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
|
4
|
de la Puente-Aldea J, Lopez-Llanos O, Horrillo D, Marcos-Sanchez H, Sanz-Ballesteros S, Franco R, Jaisser F, Senovilla L, Palacios-Ramirez R. Mineralocorticoid Receptor and Sleep Quality in Chronic Kidney Disease. Int J Mol Sci 2024; 25:12320. [PMID: 39596384 PMCID: PMC11594958 DOI: 10.3390/ijms252212320] [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: 09/09/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
The classical function of the mineralocorticoid receptor (MR) is to maintain electrolytic homeostasis and control extracellular volume and blood pressure. The MR is expressed in the central nervous system (CNS) and is involved in the regulation of the hypothalamic-pituitary-adrenal (HPA) axis as well as sleep physiology, playing a role in the non-rapid eye movement (NREM) phase of sleep. Some patients with psychiatric disorders have very poor sleep quality, and a relationship between MR dysregulation and this disorder has been found in them. In addition, the MR is involved in the regulation of the renal peripheral clock. One of the most common comorbidities observed in patients with chronic kidney disease (CKD) is poor sleep quality. Patients with CKD experience sleep disturbances, including reduced sleep duration, sleep fragmentation, and insomnia. To date, no studies have specifically investigated the relationship between MR activation and CKD-associated sleep disturbances. However, in this review, we analyzed the environment that occurs in CKD and proposed two MR-related mechanisms that may be responsible for these sleep disturbances: the circadian clock disruption and the high levels of MR agonist observed in CKD.
Collapse
Affiliation(s)
- Juan de la Puente-Aldea
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid—CSIC, 47003 Valladolid, Spain; (J.d.l.P.-A.); (O.L.-L.); (L.S.)
| | - Oscar Lopez-Llanos
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid—CSIC, 47003 Valladolid, Spain; (J.d.l.P.-A.); (O.L.-L.); (L.S.)
| | - Daniel Horrillo
- Facultad de ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcon, Spain; (D.H.); (R.F.)
| | | | | | - Raquel Franco
- Facultad de ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcon, Spain; (D.H.); (R.F.)
| | - Frederic Jaisser
- INSERM U1166, Team Diabetes, Metabolic Diseases and Comorbidities, Sorbonne Université, 75013 Paris, France;
- INSERM UMR 1116, Centre d’Investigations Cliniques-Plurithématique 1433, Université de Lorraine, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, 54500 Nancy, France
| | - Laura Senovilla
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid—CSIC, 47003 Valladolid, Spain; (J.d.l.P.-A.); (O.L.-L.); (L.S.)
- INSERM U1138, Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Sorbonne Université, Institut Universitaire de France, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
| | - Roberto Palacios-Ramirez
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid—CSIC, 47003 Valladolid, Spain; (J.d.l.P.-A.); (O.L.-L.); (L.S.)
| |
Collapse
|
5
|
Song QX, Suadicani SO, Negoro H, Jiang HH, Jabr R, Fry C, Xue W, Damaser MS. Disruption of circadian rhythm as a potential pathogenesis of nocturia. Nat Rev Urol 2024:10.1038/s41585-024-00961-0. [PMID: 39543359 DOI: 10.1038/s41585-024-00961-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 11/17/2024]
Abstract
Increasing evidence suggested the multifactorial nature of nocturia, but the true pathogenesis of this condition still remains to be elucidated. Contemporary clinical medications are mostly symptom based, aimed at either reducing nocturnal urine volume or targeting autonomic receptors within the bladder to facilitate urine storage. The day-night switch of the micturition pattern is controlled by circadian clocks located both in the central nervous system and in the peripheral organs. Arousal threshold and secretion of melatonin and vasopressin increase at night-time to achieve high-quality sleep and minimize nocturnal urine production. In response to the increased vasopressin, the kidney reduces the glomerular filtration rate and facilitates the reabsorption of water. Synchronously, in the bladder, circadian oscillation of crucial molecules occurs to reduce afferent sensory input and maintain sufficient bladder capacity during the night sleep period. Thus, nocturia might occur as a result of desynchronization in one or more of these circadian regulatory mechanisms. Disrupted rhythmicity of the central nervous system, kidney and bladder (known as the brain-kidney-bladder circadian axis) contributes to the pathogenesis of nocturia. Novel insights into the chronobiological nature of nocturia will be crucial to promote a revolutionary shift towards effective therapeutics targeting the realignment of the circadian rhythm.
Collapse
Affiliation(s)
- Qi-Xiang Song
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sylvia O Suadicani
- Department of Urology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hiromitsu Negoro
- Department of Urology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hai-Hong Jiang
- Department of Urology and Andrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rita Jabr
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Christopher Fry
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Wei Xue
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Margot S Damaser
- Department of Biomedical Engineering, Lerner Research Institute and Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
6
|
Dutta P, Layton AT. Sex and circadian regulation of metabolic demands in the rat kidney: A modeling analysis. PLoS One 2024; 19:e0293419. [PMID: 39018272 PMCID: PMC11253979 DOI: 10.1371/journal.pone.0293419] [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: 04/14/2023] [Accepted: 10/12/2023] [Indexed: 07/19/2024] Open
Abstract
Renal hemodynamics, renal transporter expression levels, and urine excretion exhibit circadian variations. Disruption of these diurnal patterns is associated with the pathophysiology of hypertension and chronic kidney disease. Renal hemodynamics determines oxygen delivery, whereas renal transport and metabolism determines oxygen consumption; the balance between them yields renal oxygenation which also demonstrates 24-h periodicity. Another notable modulator of kidney function is sex, which has impacts on renal hemodynamics and transport function that are regulated by as well as independent of the circadian clock. The goal of this study was to investigate the diurnal and sexual variations in renal oxygen consumption and oxygenation. For this purpose, we developed computational models of rat kidney function that represent sexual dimorphism and circadian variation in renal hemodynamics and transporter activities. Model simulations predicted substantial differences in tubular Na+ transport and oxygen consumption among different nephron segments. We also simulated the effect of loop diuretics, which are used in the treatment of renal hypoxia, on medullary oxygen tension. Our model predicted a significantly higher effect of loop diuretics on medullary oxygenation in female rats compared to male rats and when administered during the active phase.
Collapse
Affiliation(s)
- Pritha Dutta
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Anita T. Layton
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
- Cheriton School of Computer Science, University of Waterloo, Waterloo, Ontario, Canada
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
7
|
Bartman CM, Nesbitt L, Lee KK, Khalfaoui L, Fang Y, Pabelick CM, Prakash YS. BMAL1 sex-specific effects in the neonatal mouse airway exposed to moderate hyperoxia. Physiol Rep 2024; 12:e16122. [PMID: 38942729 PMCID: PMC11213646 DOI: 10.14814/phy2.16122] [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: 03/05/2024] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 06/30/2024] Open
Abstract
Supplemental O2 (hyperoxia) is a critical intervention for premature infants (<34 weeks) but consequently is associated with development of bronchial airway hyperreactivity (AHR) and asthma. Clinical practice shifted toward the use of moderate hyperoxia (<60% O2), but risk for subsequent airway disease remains. In mouse models of moderate hyperoxia, neonatal mice have increased AHR with effects on airway smooth muscle (ASM), a cell type involved in airway tone, bronchodilation, and remodeling. Understanding mechanisms by which moderate O2 during the perinatal period initiates sustained airway changes is critical to drive therapeutic advancements toward treating airway diseases. We propose that cellular clock factor BMAL1 is functionally important in developing mouse airways. In adult mice, cellular clocks target pathways highly relevant to asthma pathophysiology and Bmal1 deletion increases inflammatory response, worsens lung function, and impacts survival outcomes. Our understanding of BMAL1 in the developing lung is limited, but our previous findings show functional relevance of clocks in human fetal ASM exposed to O2. Here, we characterize Bmal1 in our established mouse neonatal hyperoxia model. Our data show that Bmal1 KO deleteriously impacts the developing lung in the context of O2 and these data highlight the importance of neonatal sex in understanding airway disease.
Collapse
Affiliation(s)
- Colleen M. Bartman
- Department of Anesthesiology and Perioperative MedicineMayo ClinicRochesterMinnesotaUSA
| | - Lisa Nesbitt
- Department of Anesthesiology and Perioperative MedicineMayo ClinicRochesterMinnesotaUSA
| | - Kenge K. Lee
- Department of Anesthesiology and Perioperative MedicineMayo ClinicRochesterMinnesotaUSA
| | - Latifa Khalfaoui
- Department of Anesthesiology and Perioperative MedicineMayo ClinicRochesterMinnesotaUSA
| | - Yun‐Hua Fang
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Christina M. Pabelick
- Department of Anesthesiology and Perioperative MedicineMayo ClinicRochesterMinnesotaUSA
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Y. S. Prakash
- Department of Anesthesiology and Perioperative MedicineMayo ClinicRochesterMinnesotaUSA
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
8
|
Dominguez Rieg JA, Rieg T. New functions and roles of the Na +-H +-exchanger NHE3. Pflugers Arch 2024; 476:505-516. [PMID: 38448727 DOI: 10.1007/s00424-024-02938-9] [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: 02/05/2024] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
The sodium/proton exchanger isoform 3 (NHE3) is expressed in the intestine and the kidney, where it contributes to hydrogen secretion and sodium (re)absorption. The roles of this transporter have been studied by the use of the respective knockout mice and by using pharmacological inhibitors. Whole-body NHE3 knockout mice suffer from a high mortality rate (with only ∼30% of mice surviving into adulthood), and based on the expression of NHE3 in both intestine and kidney, some conclusions that were originally derived were based on this rather complex phenotype. In the last decade, more refined models have been developed that added temporal and spatial control of NHE3 expression. For example, novel mouse models have been developed with a knockout of NHE3 in intestinal epithelial cells, tubule/collecting duct of the kidney, proximal tubule of the kidney, and thick ascending limb of the kidney. These refined models have significantly contributed to our understanding of the role of NHE3 in a tissue/cell type-specific manner. In addition, tenapanor was developed, which is a non-absorbable, intestine-specific NHE3 inhibitor. In rat and human studies, tenapanor lowered intestinal Pi uptake and was effective in lowering plasma Pi levels in patients on hemodialysis. Of note, diarrhea is seen as a side effect of tenapanor (with its indication for the treatment of constipation) and in intestine-specific NHE3 knockout mice; however, effects on plasma Pi were not supported by this mouse model which showed enhanced and not reduced intestinal Pi uptake. Further studies indicated that the gut microbiome in mice lacking intestinal NHE3 resembles an intestinal environment favoring the competitive advantage of inflammophilic over anti-inflammatory species, something similar seen in patients with inflammatory bowel disease. This review will highlight recent developments and summarize newly gained insight from these refined models.
Collapse
Affiliation(s)
- Jessica A Dominguez Rieg
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
- James A. Haley Veterans' Hospital, Tampa, FL, 33612, USA
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, 33602, USA
| | - Timo Rieg
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.
- James A. Haley Veterans' Hospital, Tampa, FL, 33612, USA.
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, 33602, USA.
| |
Collapse
|
9
|
Lee DY, Jung I, Park SY, Yu JH, Seo JA, Kim KJ, Kim NH, Yoo HJ, Kim SG, Choi KM, Baik SH, Kim NH. Attention to Innate Circadian Rhythm and the Impact of Its Disruption on Diabetes. Diabetes Metab J 2024; 48:37-52. [PMID: 38173377 PMCID: PMC10850272 DOI: 10.4093/dmj.2023.0193] [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: 06/21/2023] [Accepted: 10/16/2023] [Indexed: 01/05/2024] Open
Abstract
Novel strategies are required to reduce the risk of developing diabetes and/or clinical outcomes and complications of diabetes. In this regard, the role of the circadian system may be a potential candidate for the prevention of diabetes. We reviewed evidence from animal, clinical, and epidemiological studies linking the circadian system to various aspects of the pathophysiology and clinical outcomes of diabetes. The circadian clock governs genetic, metabolic, hormonal, and behavioral signals in anticipation of cyclic 24-hour events through interactions between a "central clock" in the suprachiasmatic nucleus and "peripheral clocks" in the whole body. Currently, circadian rhythmicity in humans can be subjectively or objectively assessed by measuring melatonin and glucocorticoid levels, core body temperature, peripheral blood, oral mucosa, hair follicles, rest-activity cycles, sleep diaries, and circadian chronotypes. In this review, we summarized various circadian misalignments, such as altered light-dark, sleep-wake, rest-activity, fasting-feeding, shift work, evening chronotype, and social jetlag, as well as mutations in clock genes that could contribute to the development of diabetes and poor glycemic status in patients with diabetes. Targeting critical components of the circadian system could deliver potential candidates for the treatment and prevention of type 2 diabetes mellitus in the future.
Collapse
Affiliation(s)
- Da Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Inha Jung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - So Young Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ji Hee Yu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ji A Seo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Kyeong Jin Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nam Hoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Hye Jin Yoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sin Gon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sei Hyun Baik
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nan Hee Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
- BK21 FOUR R&E Center for Learning Health Systems, Korea University, Seoul, Korea
| |
Collapse
|
10
|
Crislip GR, Costello HM, Juffre A, Cheng KY, Lynch IJ, Johnston JG, Drucker CB, Bratanatawira P, Agarwal A, Mendez VM, Thelwell RS, Douma LG, Wingo CS, Alli AA, Scindia YM, Gumz ML. Male kidney-specific BMAL1 knockout mice are protected from K +-deficient, high-salt diet-induced blood pressure increases. Am J Physiol Renal Physiol 2023; 325:F656-F668. [PMID: 37706232 PMCID: PMC10874679 DOI: 10.1152/ajprenal.00126.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/22/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023] Open
Abstract
The circadian clock protein basic helix-loop-helix aryl hydrocarbon receptor nuclear translocator-like protein 1 (BMAL1) is a transcription factor that impacts kidney function, including blood pressure (BP) control. Previously, we have shown that male, but not female, kidney-specific cadherin Cre-positive BMAL1 knockout (KS-BMAL1 KO) mice exhibit lower BP compared with littermate controls. The goal of this study was to determine the BP phenotype and immune response in male KS-BMAL1 KO mice in response to a low-K+ high-salt (LKHS) diet. BP, renal inflammatory markers, and immune cells were measured in male mice following an LKHS diet. Male KS-BMAL1 KO mice had lower BP following the LKHS diet compared with control mice, yet their circadian rhythm in pressure remained unchanged. Additionally, KS-BMAL1 KO mice exhibited lower levels of renal proinflammatory cytokines and immune cells following the LKHS diet compared with control mice. KS-BMAL1 KO mice were protected from the salt-sensitive hypertension observed in control mice and displayed an attenuated immune response following the LKHS diet. These data suggest that BMAL1 plays a role in driving the BP increase and proinflammatory environment that occurs in response to an LKHS diet.NEW & NOTEWORTHY We show here, for the first time, that kidney-specific BMAL1 knockout mice are protected from blood pressure (BP) increases and immune responses to a salt-sensitive diet. Other kidney-specific BMAL1 knockout models exhibit lower BP phenotypes under basal conditions. A salt-sensitive diet exacerbates this genotype-specific BP response, leading to fewer proinflammatory cytokines and immune cells in knockout mice. These data demonstrate the importance of distal segment BMAL1 in BP and immune responses to a salt-sensitive environment.
Collapse
Affiliation(s)
- 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 Disease, University of Florida, Gainesville, Florida, United States
| | - 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 Disease, University of Florida, Gainesville, Florida, United States
| | - Alexandria Juffre
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, Florida, United States
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, United States
| | - Kit-Yan Cheng
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - I Jeanette Lynch
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
- Department of Research, North Florida/South Georgia Veterans Health System, Gainesville, Florida, United States
| | - Jermaine G Johnston
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, Florida, United States
- Department of Research, North Florida/South Georgia Veterans Health System, Gainesville, Florida, United States
| | - Charles B Drucker
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Phillip Bratanatawira
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
| | - Annanya Agarwal
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, United States
| | - Victor M Mendez
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
| | - Ryanne S Thelwell
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
| | - Lauren G Douma
- 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
| | - Charles S Wingo
- 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 Research, North Florida/South Georgia Veterans Health System, Gainesville, Florida, United States
| | - Abdel A Alli
- 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 Disease, University of Florida, Gainesville, Florida, United States
| | - Yogesh M Scindia
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, 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 Disease, University of Florida, Gainesville, Florida, United States
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
11
|
Bingham MA, Neijman K, Yang CR, Aponte A, Mak A, Kikuchi H, Jung HJ, Poll BG, Raghuram V, Park E, Chou CL, Chen L, Leipziger J, Knepper MA, Dona M. Circadian gene expression in mouse renal proximal tubule. Am J Physiol Renal Physiol 2023; 324:F301-F314. [PMID: 36727945 PMCID: PMC9988533 DOI: 10.1152/ajprenal.00231.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Circadian variability in kidney function is well recognized but is often ignored as a potential confounding variable in physiological experiments. Here, we have created a data resource consisting of expression levels for mRNA transcripts in microdissected proximal tubule segments from mice as a function of the time of day. Small-sample RNA sequencing was applied to microdissected S1 proximal convoluted tubules and S2 proximal straight tubules. After stringent filtering, the data were analyzed using JTK-Cycle to detect periodicity. The data set is provided as a user-friendly webpage at https://esbl.nhlbi.nih.gov/Databases/Circadian-Prox2/. In proximal convoluted tubules, 234 transcripts varied in a circadian manner (4.0% of the total). In proximal straight tubules, 334 transcripts varied in a circadian manner (5.3%). Transcripts previously known to be associated with corticosteroid action and with increased flow were found to be overrepresented among circadian transcripts peaking during the "dark" portion of the day [zeitgeber time (ZT)14-22], corresponding to peak levels of corticosterone and glomerular filtration rate in mice. To ask whether there is a time-of-day dependence of protein abundances in the kidney, we carried out LC-MS/MS-based proteomics in whole mouse kidneys at ZT12 and ZT0. The full data set (n = 6,546 proteins) is available at https://esbl.nhlbi.nih.gov/Databases/Circadian-Proteome/. Overall, 293 proteins were differentially expressed between ZT12 and ZT0 (197 proteins greater at ZT12 and 96 proteins greater at ZT0). Among the regulated proteins, only nine proteins were found to be periodic in the RNA-sequencing analysis, suggesting a high level of posttranscriptional regulation of protein abundances.NEW & NOTEWORTHY Circadian variation in gene expression can be an important determinant in the regulation of kidney function. The authors used RNA-sequencing transcriptomics and LC-MS/MS-based proteomics to identify gene products expressed in a periodic manner. The data were used to construct user-friendly web resources.
Collapse
Affiliation(s)
- Molly A Bingham
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Kim Neijman
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Chin-Rang Yang
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Angel Aponte
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Angela Mak
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Hiroaki Kikuchi
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Hyun Jun Jung
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Brian G Poll
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Viswanathan Raghuram
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Euijung Park
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Chung-Lin Chou
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Lihe Chen
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Jens Leipziger
- Department of Biomedicine, Physiology, Aarhus University, Aarhus, Denmark
| | - Mark A Knepper
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Margo Dona
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Dutta P, Sadria M, Layton AT. Influence of administration time and sex on natriuretic, diuretic, and kaliuretic effects of diuretics. Am J Physiol Renal Physiol 2023; 324:F274-F286. [PMID: 36701479 DOI: 10.1152/ajprenal.00296.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Sex differences in renal function and blood pressure have been widely described across many species. Blood pressure dips during sleep and peaks in the early morning. Similarly, glomerular filtration rate, filtered electrolyte loads, urine volume, and urinary excretion all exhibit notable diurnal rhythms, which reflect, in part, the regulation of renal transporter proteins by circadian clock genes. That regulation is sexually dimorphic; as such, sex and time of day are not two independent regulators of kidney function and blood pressure. The objective of the present study was to assess the effect of sex and administration time on the natriuretic and diuretic effects of loop, thiazide, and K+-sparing diuretics, which are common treatments for hypertension. Loop diuretics inhibit Na+-K+-2Cl- cotransporters on the apical membrane of the thick ascending limb, thiazide diuretics inhibit Na+-Cl- cotransporters on the distal convoluted tubule, and K+-sparing diuretics inhibit epithelial Na+ channels on the connecting tubule and collecting duct. We simulated Na+ transporter inhibition using sex- and time-of-day-specific computational models of mouse kidney function. The simulation results highlighted significant sex and time-of-day differences in the drug response. Loop diuretics induced larger natriuretic and diuretic effects during the active phase. The natriuretic and diuretic effects of thiazide diuretics exhibited sex and time-of-day differences, whereas these effects of K+-sparing diuretics exhibited a significant time-of-day difference in females only. The kaliuretic effect depended on the type of diuretics and time of administration. The present computational models can be a useful tool in chronotherapy, to tailor drug administration time to match the body's diurnal rhythms to optimize the drug effect.NEW & NOTEWORTHY Sex influences cardiovascular disease, and the timing of onset of acute cardiovascular events exhibits circadian rhythms. Kidney function also exhibits sex differences and circadian rhythms. How do the natriuretic and diuretic effects of diuretics, a common treatment for hypertension that targets the kidneys, differ between the sexes? And how do these effects vary during the day? To answer these questions, we conducted computer simulations to assess the effects of loop, thiazide, and K+-sparing diuretics.
Collapse
Affiliation(s)
- Pritha Dutta
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Mehrshad Sadria
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Anita T Layton
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada.,Cheriton School of Computer Science, University of Waterloo, Waterloo, Ontario, Canada.,Department of Biology, University of Waterloo, Waterloo, Ontario, Canada.,School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
13
|
Wang J, She Q, Du J. Dapagliflozin attenuates myocardial remodeling in hypertension by activating the circadian rhythm signaling pathway. Arch Pharm Res 2023; 46:117-130. [PMID: 36729273 DOI: 10.1007/s12272-023-01430-9] [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: 08/30/2022] [Accepted: 01/27/2023] [Indexed: 02/03/2023]
Abstract
Sodium-glucose cotransporter 2 inhibitor (SGLT2i) is a new kind of antidiabetic drug which has shown beneficial effects in reducing heart failure-related hospitalization and cardiovascular-related mortality. The mechanisms are complicated. Our study aimed to investigate the effects of dapagliflozin on the myocardium of spontaneously hypertensive rats (SHRs) without heart failure. Wistar-Kyoto rats were used as normal controls. SHRs were randomly divided into the SHR group and the -treated group. After 8 weeks of dapagliflozin treatment, the morphology of heart tissues was examined. The mRNA expression profiles were identified via RNA sequencing (RNA-Seq). Various analysis methods were used to find the differentially expressed genes (DEGs) to predict gene function and coexpression. After dapagliflozin treatment, systolic blood pressure was significantly reduced compared with that in the SHR group. Myocardial remodeling was ameliorated compared with that in the SHR group. After dapagliflozin intervention, 75 DEGs (|log2-fold change | > 0 and Q value < 0.05) were identified in the heart tissues compared to the SHR group. Quantitative real-time PCR analysis confirmed that the expression of the circadian rhythm genes Per3, Bhlhe41, and Nr1d1 was significantly upregulated, while the results were coincident with the RNA-Seq results. Dapagliflozin may effectively inhibit myocardial remodeling and regulate blood pressure. The mechanisms may be related to the activation of the circadian rhythm signaling pathway.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
14
|
Preston R, Meng QJ, Lennon R. The dynamic kidney matrisome - is the circadian clock in control? Matrix Biol 2022; 114:138-155. [PMID: 35569693 DOI: 10.1016/j.matbio.2022.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/21/2022] [Accepted: 05/10/2022] [Indexed: 02/06/2023]
Abstract
The circadian clock network in mammals is responsible for the temporal coordination of numerous physiological processes that are necessary for homeostasis. Peripheral tissues demonstrate circadian rhythmicity and dysfunction of core clock components has been implicated in the pathogenesis of diseases that are characterized by abnormal extracellular matrix, such as fibrosis (too much disorganized matrix) and tissue breakdown (too little matrix). Kidney disease is characterized by proteinuria, which along with the rate of filtration, displays robust circadian oscillation. Clinical observation and mouse studies suggest the presence of 24 h kidney clocks responsible for circadian oscillation in kidney function. Recent experimental evidence has also revealed that cell-matrix interactions and the biomechanical properties of extracellular matrix have key roles in regulating peripheral circadian clocks and this mechanism appears to be cell- and tissue-type specific. Thus, establishing a temporally resolved kidney matrisome may provide a useful tool for studying the two-way interactions between the extracellular matrix and the intracellular time-keeping mechanisms in this critical niche tissue. This review summarizes the latest genetic and biochemical evidence linking kidney physiology and disease to the circadian system with a particular focus on the extracellular matrix. We also review the experimental approaches and methodologies required to dissect the roles of circadian pathways in specific tissues and outline the translational aspects of circadian biology, including how circadian medicine could be used for the treatment of kidney disease.
Collapse
Affiliation(s)
- Rebecca Preston
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Qing-Jun Meng
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK.
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK; Department of Pediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK.
| |
Collapse
|
15
|
Layton AT, Gumz ML. Sex differences in circadian regulation of kidney function of the mouse. Am J Physiol Renal Physiol 2022; 323:F675-F685. [PMID: 36264883 PMCID: PMC11905794 DOI: 10.1152/ajprenal.00227.2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/05/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022] Open
Abstract
Kidney function is regulated by the circadian clock. Not only do glomerular filtration rate and urinary excretion oscillate during the day, but the expressions of several renal transporter proteins also exhibit circadian rhythms. Interestingly, the circadian regulation of these transporters appears to be sexually dimorphic. Thus, the goal of the present study was to investigate the mechanisms by which the kidney function of the mouse is modulated by sex and time of day. To accomplish this, we developed the first computational models of epithelial water and solute transport along the mouse nephrons that represent the effects of sex and the circadian clock on renal hemodynamics and transporter activity. We conducted simulations to study how the circadian control of renal transport genes affects overall kidney function and how that process differs between male and female mice. Simulation results predicted that tubular transport differs substantially among segments, with relative variations in water and Na+ reabsorption along the proximal tubules and thick ascending limb tracking that of glomerular filtration rate. In contrast, relative variations in distal segment transport were much larger, with Na+ reabsorption almost doubling during the active phase. Oscillations in Na+ transport drive K+ transport variations in the opposite direction. Model simulations of basic helix-loop-helix ARNT like 1 (BMAL1) knockout mice predicted a significant reduction in net Na+ reabsorption along the distal segments in both sexes, but more so in males than in females. This can be attributed to the reduction of mean epithelial Na+ channel activity in males only, a sex-specific effect that may lead to a reduction in blood pressure in BMAL1-null males.NEW & NOTEWORTHY How does the circadian control of renal transport genes affect overall kidney function, and how does that process differ between male and female mice? How does the differential circadian regulation of the expression levels of key transporter genes impact the transport processes along different nephron segments during the day? And how do those effects differ between males and females? We built computational models of mouse kidney function to answer these questions.
Collapse
Affiliation(s)
- Anita T Layton
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
- Cheriton School of Computer Science, University of Waterloo, Waterloo, Ontario, Canada
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| | - Michelle L Gumz
- Department of Physiology and Aging, University of Florida, Gainesville, Florida
| |
Collapse
|
16
|
Costello HM, Johnston JG, Juffre A, Crislip GR, Gumz ML. Circadian clocks of the kidney: function, mechanism, and regulation. Physiol Rev 2022; 102:1669-1701. [PMID: 35575250 PMCID: PMC9273266 DOI: 10.1152/physrev.00045.2021] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/22/2022] Open
Abstract
An intrinsic cellular circadian clock is located in nearly every cell of the body. The peripheral circadian clocks within the cells of the kidney contribute to the regulation of a variety of renal processes. In this review, we summarize what is currently known regarding the function, mechanism, and regulation of kidney clocks. Additionally, the effect of extrarenal physiological processes, such as endocrine and neuronal signals, on kidney function is also reviewed. Circadian rhythms in renal function are an integral part of kidney physiology, underscoring the importance of considering time of day as a key biological variable. The field of circadian renal physiology is of tremendous relevance, but with limited physiological and mechanistic information on the kidney clocks this is an area in need of extensive investigation.
Collapse
Affiliation(s)
- 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
| | - Jermaine G Johnston
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcom Randall Department of 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
| | - 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
| | - 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
- North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, 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
|
17
|
Onuma S, Kinoshita S, Shimba S, Ozono K, Michigami T, Kawai M. The Lack of Bmal1, a Core Clock Gene, in the Intestine Decreases Glucose Absorption in Mice. Endocrinology 2022; 163:6651710. [PMID: 35904419 DOI: 10.1210/endocr/bqac119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Indexed: 11/19/2022]
Abstract
The circadian clock network is an evolutionarily conserved system that regulates systemic metabolism, such as glucose homeostasis. Intestinal tissue is a pivotal organ for the regulation of glucose metabolism, mainly via glucose absorption into the circulation; however, the significance of the intestinal circadian clock network for glucose metabolism remains largely unclear. We herein utilized a mouse model in which Bmal1, a core clock gene, was deleted in an intestine-specific manner (Bmal1Int-/- mice) and demonstrated a rhythmic expression of Sglt1 with its peak at zeitgeber time (ZT) 10.7 ± 2.8 in control mice, whereas this was lost in Bmal1Int-/- mice. Mechanistically, chromatin immunoprecipitation analysis revealed rhythmic binding of CLOCK to the E-box elements in the Sglt1 gene in control mice; however, this was absent in Bmal1Int-/- mice. Accordingly, SGLT1 protein levels were decreased during the dark phase in Bmal1Int-/- mice and this was associated with impaired glucose absorption, leading to a decline in hepatic glycogen levels at ZT4, which was restored by ingestion of high-sucrose water. Additionally, when mice were starved from ZT0, greater expression of the lipolysis-related gene Pnpla2 was observed in adipose tissue of Bmal1Int-/- mice, and this was not noted when glycogen storage was restored by high-sucrose water prior to fasting, suggesting that higher Pnpla2 expression in Bmal1Int-/- mice was likely caused by lower glycogen storage. These results indicate that disruption of the intestinal circadian clock system impairs glucose absorption in the intestine and affects systemic glucose homeostasis.
Collapse
Affiliation(s)
- Shinsuke Onuma
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Izumi, Osaka 594-1101, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Saori Kinoshita
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Izumi, Osaka 594-1101, Japan
| | - Shigeki Shimba
- Department of Health Science, School of Pharmacy, Nihon University, Funabashi, Chiba 274-8555, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Izumi, Osaka 594-1101, Japan
| | - Masanobu Kawai
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Izumi, Osaka 594-1101, Japan
| |
Collapse
|
18
|
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: 19] [Impact Index Per Article: 6.3] [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
|
19
|
Itoh H, Tanaka M. “Greedy Organs Hypothesis” for sugar and salt in the pathophysiology of non-communicable diseases in relation to sodium-glucose co-transporters in the intestines and the kidney. Metabol Open 2022; 13:100169. [PMID: 35198947 PMCID: PMC8844901 DOI: 10.1016/j.metop.2022.100169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 11/05/2022] Open
Abstract
Deposition of visceral fat and insulin resistance play central role in the development of non-communicable diseases (NCDs) including obesity, hypertension and type 2 diabetes. However, we shed more light upon the intestines and the kidney as a strong driver of NCDs. Based upon unexpected outcomes of clinical trials using sodium-glucose cotransporter (SGLT) 2 inhibitors to demonstrate their actions for not only body weight reduction and blood glucose fall but also remarkable cardiorenal protection, we speculate that hyperfunction of the intestines and the kidney is one of critical contributing factors for initiation of NCDs. By detecting high amount of glucose and sodium chloride around them by sweet/salt taste sensors, the intestines and the kidney are designed to (re)absorb these nutrients by up-regulating SGLT1 or SGLT2. We designate these hyperfunctioning organs for nutrient uptake as “greedy organs”. The greedy organs can induce NCDs (“greedy organ hypothesis”). SGLTs are regulated by glucose and sodium chloride, and SGLTs or other genes can be “greedy genes.” Regulating factors for greedy organs are renin-angiotensin system, renal sympathetic nervous activity, gut inflammation/microbiota or oxidative stress. Mitigation of organ greediness by SGLT2 inhibitors, ketone bodies, bariatric surgery, and regular lifestyle to keep rhythmicity of biological clock are promising. We propose the concept of “Greedy Organs” hypothesis as a possible cause of NCDs. Clinical implication of greedy kidney is supported by the effect of SGLT2 inhibitors. The significance of greedy intestines is suggested by the effect of bariatric surgery. The intestines and kidney become hyperactive through upregulation of SGLT1 or 2. To mitigate “greedy organs” should be a promising strategy against NCDs.
Collapse
|
20
|
Obata F, Ozuru R, Tsuji T, Matsuba T, Fujii J. Stx2 Induces Differential Gene Expression and Disturbs Circadian Rhythm Genes in the Proximal Tubule. Toxins (Basel) 2022; 14:toxins14020069. [PMID: 35202097 PMCID: PMC8874938 DOI: 10.3390/toxins14020069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/04/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) causes proximal tubular defects in the kidney. However, factors altered by Shiga toxin (Stx) within the proximal tubules are yet to be shown. We determined Stx receptor Gb3 in murine and human kidneys and confirmed the receptor expression in the proximal tubules. Stx2-injected mouse kidney tissues and Stx2-treated human primary renal proximal tubular epithelial cell (RPTEC) were collected and microarray analysis was performed. We compared murine kidney and RPTEC arrays and selected common 58 genes that are differentially expressed vs. control (0 h, no toxin-treated). We found that the most highly expressed gene was GDF15, which may be involved in Stx2-induced weight loss. Genes associated with previously reported Stx2 activities such as src kinase Yes phosphorylation pathway activation, unfolded protein response (UPR) and ribotoxic stress response (RSR) showed differential expressions. Moreover, circadian clock genes were differentially expressed, suggesting Stx2-induced renal circadian rhythm disturbance. Circadian rhythm-regulated proximal tubular Na+-glucose transporter SGLT1 (SLC5A1) was down-regulated, indicating proximal tubular functional deterioration, and mice developed glucosuria confirming proximal tubular dysfunction. Stx2 alters gene expression in murine and human proximal tubules through known activities and newly investigated circadian rhythm disturbance, which may result in proximal tubular dysfunctions.
Collapse
Affiliation(s)
- Fumiko Obata
- Division of Bacteriology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, 86 Nishicho, Yonago 683-8503, Japan; (T.T.); (J.F.)
- Correspondence:
| | - Ryo Ozuru
- Department of Microbiology and Immunology, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan;
| | - Takahiro Tsuji
- Division of Bacteriology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, 86 Nishicho, Yonago 683-8503, Japan; (T.T.); (J.F.)
| | - Takashi Matsuba
- Division of Infectious Disease Control and Prevention, Department of Animal Pharmaceutical Science, School of Pharmaceutical Science, Kyusyu University of Health and Welfare, 1714-1 Yoshino-machi, Nobeoka 882-8508, Japan;
| | - Jun Fujii
- Division of Bacteriology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, 86 Nishicho, Yonago 683-8503, Japan; (T.T.); (J.F.)
| |
Collapse
|
21
|
Ansermet C, Centeno G, Bignon Y, Ortiz D, Pradervand S, Garcia A, Menin L, Gachon F, Yoshihara HA, Firsov D. Dysfunction of the circadian clock in the kidney tubule leads to enhanced kidney gluconeogenesis and exacerbated hyperglycemia in diabetes. Kidney Int 2021; 101:563-573. [PMID: 34838539 DOI: 10.1016/j.kint.2021.11.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 11/01/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022]
Abstract
The circadian clock is a ubiquitous molecular time-keeping mechanism which synchronizes cellular, tissue, and systemic biological functions with 24-hour environmental cycles. Local circadian clocks drive cell type- and tissue-specific rhythms and their dysregulation has been implicated in pathogenesis and/or progression of a broad spectrum of diseases. However, the pathophysiological role of intrinsic circadian clocks in the kidney of diabetics remains unknown. To address this question, we induced type I diabetes with streptozotocin in mice devoid of the circadian transcriptional regulator BMAL1 in podocytes (cKOp mice) or in the kidney tubule (cKOt mice). There was no association between dysfunction of the circadian clock and the development of diabetic nephropathy in cKOp and cKOt mice with diabetes. However, cKOt mice with diabetes exhibited exacerbated hyperglycemia, increased fractional excretion of glucose in the urine, enhanced polyuria, and a more pronounced kidney hypertrophy compared to streptozotocin-treated control mice. mRNA and protein expression analyses revealed substantial enhancement of the gluconeogenic pathway in kidneys of cKOt mice with diabetes as compared to diabetic control mice. Transcriptomic analysis along with functional analysis of cKOt mice with diabetes identified changes in multiple mechanisms directly or indirectly affecting the gluconeogenic pathway. Thus, we demonstrate that dysfunction of the intrinsic kidney tubule circadian clock can aggravate diabetic hyperglycemia via enhancement of gluconeogenesis in the kidney proximal tubule and further highlight the importance of circadian behavior in patients with diabetes.
Collapse
Affiliation(s)
- Camille Ansermet
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Gabriel Centeno
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Yohan Bignon
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Daniel Ortiz
- Mass Spectrometry Service, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Sylvain Pradervand
- Genomic Technologies Facility, University of Lausanne, Lausanne, Switzerland
| | - Andy Garcia
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Laure Menin
- Mass Spectrometry Service, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Frédéric Gachon
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland; Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Hikari Ai Yoshihara
- Institute of Physics, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Dmitri Firsov
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
22
|
Soliman RH, Pollock DM. Circadian Control of Sodium and Blood Pressure Regulation. Am J Hypertens 2021; 34:1130-1142. [PMID: 34166494 PMCID: PMC9526808 DOI: 10.1093/ajh/hpab100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 05/12/2021] [Accepted: 06/23/2021] [Indexed: 01/26/2023] Open
Abstract
The attention for the control of dietary risk factors involved in the development of hypertension, includes a large effort on dietary salt restrictions. Ample studies show the beneficial role of limiting dietary sodium as a lifestyle modification in the prevention and management of essential hypertension. Not until the past decade or so have studies more specifically investigated diurnal variations in renal electrolyte excretion, which led us to the hypothesis that timing of salt intake may impact cardiovascular health and blood pressure regulation. Cell autonomous molecular clocks as the name implies, function independently to maintain optimum functional rhythmicity in the face of environmental stressors such that cellular homeostasis is maintained at all times. Our understanding of mechanisms influencing diurnal patterns of sodium excretion and blood pressure has expanded with the discovery of the circadian clock genes. In this review, we discuss what is known about circadian regulation of renal sodium handling machinery and its influence on blood pressure regulation, with timing of sodium intake as a potential modulator of the kidney clock.
Collapse
Affiliation(s)
- Reham H Soliman
- Section of Cardio-renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David M Pollock
- Section of Cardio-renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
23
|
Nakamoto I, Uiji S, Okata R, Endo H, Tohyama S, Nitta R, Hashimoto S, Matsushima Y, Wakimoto J, Hashimoto S, Nishiyama Y, Kanikowska D, Negoro H, Wakamura T. Diurnal rhythms of urine volume and electrolyte excretion in healthy young men under differing intensities of daytime light exposure. Sci Rep 2021; 11:13097. [PMID: 34162962 PMCID: PMC8222329 DOI: 10.1038/s41598-021-92595-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/14/2021] [Indexed: 11/17/2022] Open
Abstract
In humans, most renal functions, including urine volume and electrolyte excretions, have a circadian rhythm. Light is a strong circadian entrainment factor and daytime-light exposure is known to affect the circadian rhythm of rectal temperature (RT). The effects of daytime-light exposure on the diurnal rhythm of urinary excretion have yet to be clarified. The aim of this study was to clarify whether and how daytime exposure to bright-light affects urinary excretions. Twenty-one healthy men (21–27 years old) participated in a 4-day study involving daytime (08:00–18:00 h) exposure to two light conditions, Dim (< 50 lx) and Bright (~ 2500 lx), in a random order. During the experiment, RT was measured continuously. Urine samples were collected every 3 ~ 4 h. Compared to the Dim condition, under the Bright condition, the RT nadir time was 45 min earlier (p = 0.017) and sodium (Na), chloride (Cl), and uric acid (UA) excretion and urine volumes were greater (all p < 0.001), from 11:00 h to 13:00 h without a difference in total daily urine volume. The present results suggest that daytime bright light exposure can induce a phase shift advance in urine volume and urinary Na, Cl, and UA excretion rhythms.
Collapse
Affiliation(s)
- Isuzu Nakamoto
- Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 kawahara-cho, shogoin, sakyo-ku, Kyoto, 606-8507, Japan
| | - Sayaka Uiji
- Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 kawahara-cho, shogoin, sakyo-ku, Kyoto, 606-8507, Japan
| | - Rin Okata
- Human Health Sciences, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Hisayoshi Endo
- Human Health Sciences, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Sena Tohyama
- Human Health Sciences, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Rina Nitta
- Human Health Sciences, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Saya Hashimoto
- Human Health Sciences, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshiko Matsushima
- Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 kawahara-cho, shogoin, sakyo-ku, Kyoto, 606-8507, Japan
| | - Junko Wakimoto
- Clinical Laboratory, Kyoto University Hospital, Kyoto, Japan
| | - Seiji Hashimoto
- Clinical Laboratory, Kyoto University Hospital, Kyoto, Japan
| | | | - Dominika Kanikowska
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Tomoko Wakamura
- Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 kawahara-cho, shogoin, sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
24
|
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
|
25
|
ElDash RM, Raslan MA, Shaheen SM, Sabri NA. The effect of morning versus evening administration of empagliflozin on its pharmacokinetics and pharmacodynamics characteristics in healthy adults: a two-way crossover, non-randomised trial. F1000Res 2021; 10:321. [PMID: 34123370 PMCID: PMC8167502 DOI: 10.12688/f1000research.51114.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2021] [Indexed: 01/15/2023] Open
Abstract
Background: Empagliflozin is an SGLT2 inhibitor approved for use in patients with diabetes mellitus type 2 (DMT2) with or without other cardiovascular disease. Empagliflozin is taken once daily without rationale on the optimal timing for administration. This study aimed to determine the chronopharmacological effects of morning vs evening administration of empagliflozin (10 mg) in healthy Egyptian adults, by investigating the pharmacokinetics and pharmacodynamics parameters of empagliflozin depending on the intake time. Methods: An open label, sequential, two-way crossover trial comprised of two periods with a washout period of 7 days. All participants received a single oral dose of empagliflozin (JARDIANCE ®; 10 mg film coated tablet) in the evening, and after a seven-day washout period, the morning. Pharmacokinetics parameters (primary endpoints: t max (h), C max (ng/ml), AUC 0-t (ng.h/ml); secondary endpoints: AUC 0 to ∞(ng.h/ml)) were assessed. Method validation was done prior to injection in LC/MS/MS and samples were processed by Liquid-Liquid extraction. The pharmacodynamic profile (UGE 0-24) was determined after method validation (glucose hexokinase method). Results: T max increased by 35% in the evening phase compared to the morning phase, while C max decreased by -6.5% in the evening dose compared to the morning dose. Additionally, AUC 0 to ∞ increased in the evening phase by 8.25% compared to the morning phase. The mean cumulative amount of glucose excreted (UGE ( 0-24)) increased by 43% in the evening dose compared to the morning dose Conclusion: Despite the difference in pharmacokinetics parameters between evening and morning doses, C max, AUC 0-t, AUC 0-∞, didn't differ on the bioequivalence level. In addition, as UGE ( 0-24) didn't statistically differ, thus, we can conclude that there is no statistical significance between the morning and evening doses. Trial registration: Clinal Trials.gov, ID: NCT03895229 (registered on 29 th March 2019).
Collapse
Affiliation(s)
- Rana M. ElDash
- Pharmacy Practice Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | | | - Sara M. Shaheen
- Clinical Pharmacy Department, Faculty of Pharmacy, AinShms University, Cairo, 11566, Egypt
| | - Nagwa Ali Sabri
- Clinical Pharmacy Department, Faculty of Pharmacy, AinShms University, Cairo, 11566, Egypt
| |
Collapse
|
26
|
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
|
27
|
Wang X, Guo K, Huang B, Lin Z, Cai Z. Role of Glucose Transporters in Drug Membrane Transport. Curr Drug Metab 2020; 21:947-958. [PMID: 32778021 DOI: 10.2174/1389200221666200810125924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/12/2020] [Accepted: 06/01/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Glucose is the main energy component of cellular activities. However, as a polar molecule, glucose cannot freely pass through the phospholipid bilayer structure of the cell membrane. Thus, glucose must rely on specific transporters in the membrane. Drugs with a similar chemical structure to glucose may also be transported through this pathway. METHODS This review describes the structure, distribution, action mechanism and influencing factors of glucose transporters and introduces the natural drugs mediated by these transporters and drug design strategies on the basis of this pathway. RESULTS The glucose transporters involved in glucose transport are of two major types, namely, Na+-dependent and Na+-independent transporters. Glucose transporters can help some glycoside drugs cross the biological membrane. The transmembrane potential is influenced by the chemical structure of drugs. Glucose can be used to modify drugs and improve their ability to cross biological barriers. CONCLUSION The membrane transport mechanism of some glycoside drugs may be related to glucose transporters. Glucose modification may improve the oral bioavailability of drugs or achieve targeted drug delivery.
Collapse
Affiliation(s)
- Xin Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Kunkun Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Baolin Huang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Zimin Lin
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Zheng Cai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW Blood pressure (BP) exhibits strong diurnal variations that have been shown to be important for normal physiology and health. In this review, we highlight recent advances in both basic and clinic research on how the circadian clock affects these BP rhythms. RECENT FINDINGS Tissue-specific and inducible knockout rodent models have provided novel ways to dissect how circadian clocks regulate BP rhythms and demonstrated that loss of these rhythms is associated with the development of disease. The use of circadian-specific research protocols has translated findings from rodent models to humans, providing insight into circadian control of BP, as well as how sleep, activity, and other factors influence diurnal BP rhythms. Circadian mechanisms play an important role in the regulation of diurnal BP rhythms. Future research needs to extend these findings to clinical populations and determine the extent to which circadian factors may play a role in the development of novel treatment approaches to the management of hypertension.
Collapse
Affiliation(s)
- Megan K Rhoads
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vikhram Balagee
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - S Justin Thomas
- Department of Psychiatry, University of Alabama at Birmingham, SC1010, 1720 2nd Avenue South, Birmingham, AL, 35294-0017, USA.
| |
Collapse
|
29
|
Antihypertensive and Renal Mechanisms of SGLT2 (Sodium-Glucose Linked Transporter 2) Inhibitors. Hypertension 2020; 75:894-901. [DOI: 10.1161/hypertensionaha.119.11684] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Empaglifolzin, canagliflozin, and dapagliflozin are SGLT2 (sodium-glucose linked transporter type 2) inhibitors for treatment of type 2 diabetes mellitus that also reduce blood pressure, mortality, and cardiovascular disease and slow the loss of glomerular filtration rate. SGLT2 inhibitors inhibit the coupled reabsorption of sodium and glucose from the proximal tubules, thereby increasing renal glucose and sodium excretion, but they have more widespread renal effects, including inhibition of the sodium:proton exchanger. They increase the delivery of sodium to the loop of Henle and can thereby activate the tubuloglomerular feedback response to correct glomerular hyperfiltration. There are multiple potential mechanisms whereby these drugs lower blood pressure and preserve kidney function that are the focus of this review.
Collapse
|
30
|
Endoplasmic Reticulum Stress Signalling Induces Casein Kinase 1-Dependent Formation of Cytosolic TDP-43 Inclusions in Motor Neuron-Like Cells. Neurochem Res 2020; 45:1354-1364. [PMID: 31280399 PMCID: PMC7260270 DOI: 10.1007/s11064-019-02832-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 12/30/2022]
Abstract
Motor neuron disease (MND) is a progressive neurodegenerative disease with no effective treatment. One of the principal pathological hallmarks is the deposition of TAR DNA binding protein 43 (TDP-43) in cytoplasmic inclusions. TDP-43 aggregation occurs in both familial and sporadic MND; however, the mechanism of endogenous TDP-43 aggregation in disease is incompletely understood. This study focused on the induction of cytoplasmic accumulation of endogenous TDP-43 in the motor neuronal cell line NSC-34. The endoplasmic reticulum (ER) stressor tunicamycin induced casein kinase 1 (CK1)-dependent cytoplasmic accumulation of endogenous TDP-43 in differentiated NSC-34 cells, as seen by immunocytochemistry. Immunoblotting showed that induction of ER stress had no effect on abundance of TDP-43 or phosphorylated TDP-43 in the NP-40/RIPA soluble fraction. However, there were significant increases in abundance of TDP-43 and phosphorylated TDP-43 in the NP-40/RIPA-insoluble, urea-soluble fraction, including high molecular weight species. In all cases, these increases were lowered by CK1 inhibition. Thus ER stress signalling, as induced by tunicamycin, causes CK1-dependent phosphorylation of TDP-43 and its consequent cytosolic accumulation.
Collapse
|
31
|
Vagnerová K, Ergang P, Soták M, Balounová K, Kvapilová P, Vodička M, Pácha J. Diurnal expression of ABC and SLC transporters in jejunum is modulated by adrenalectomy. Comp Biochem Physiol C Toxicol Pharmacol 2019; 226:108607. [PMID: 31422161 DOI: 10.1016/j.cbpc.2019.108607] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 01/20/2023]
Abstract
The circadian clock system drives many physiological processes, including plasma concentration of glucocorticoids and epithelial transport of some ions and nutrients. As glucocorticoids entrain the circadian rhythms in various peripheral organs, we examined whether adrenalectomy affects the expression and circadian rhythmicity of intestinal transporters of the solute carrier (SLC) and ATP-binding cassette (ABC) families, which participate in intestinal barriers for absorption of nutrients, nonnutrients and oral drugs. The rat jejunum showed rhythmic circadian profiles of Sglt1, Pept1, Nhe3, Mdr1 and Mrp2 but not Mct1, Oct1, Octn1, Oatp1, Cnt1 and Bcrp. With the exception of Pept1 and Mct1, adrenalectomy decreased the expression of all rhythmic and arrhythmic transporters including the amplitude of Sglt1 and Nhe3 rhythms but minimally affected the phases of rhythmic transporters except of Nhe3. Similarly, adrenalectomy downregulated the expression of rhythmic (Pparα, Hlf, Pgc1α) and arrhythmic (Hnf1β, Hnf4α) transcription factors, which are known to regulate the expression of transporters. We conclude that endogenous corticosteroids have a profound effect on the expression of intestinal SLC and ABC transporters and their nuclear transcription factors. The circulating corticosteroids are necessary for maintaining upregulated expression of Sglt1, Oct1, Octn1, Oatp1, Cnt1, Nhe3, Mdr1, Bcrp, Mrp2, Pparα, Pgc1α, Hnf1β, Hnf4α and Hlf and for maintaining the high amplitude of Sglt1, Nhe3, Pparα, Pgc1α and Hlf circadian rhythms. The study demonstrates that signals from the adrenal gland are necessary for maintaining the expression of arrhythmic and rhythmic intestinal transporters and that changes in the secretion of corticosteroids associated with stress might reorganize intestinal transport barriers.
Collapse
Affiliation(s)
- Karla Vagnerová
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Peter Ergang
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Matúš Soták
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Kateřina Balounová
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic; Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Pavlína Kvapilová
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Vodička
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Jiří Pácha
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic; Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic.
| |
Collapse
|
32
|
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: 20] [Impact Index Per Article: 3.3] [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
|
33
|
Abstract
Numerous physiological functions exhibit substantial circadian oscillations. In the kidneys, renal plasma flow, the glomerular filtration rate and tubular reabsorption and/or secretion processes have been shown to peak during the active phase and decline during the inactive phase. These functional rhythms are driven, at least in part, by a self-sustaining cellular mechanism termed the circadian clock. The circadian clock controls different cellular functions, including transcription, translation and protein post-translational modifications (such as phosphorylation, acetylation and ubiquitylation) and degradation. Disruption of the circadian clock in animal models results in the loss of blood pressure control and substantial changes in the circadian pattern of water and electrolyte excretion in the urine. Kidney-specific suppression of the circadian clock in animals implicates both the intrinsic renal and the extrarenal circadian clocks in these pathologies. Alterations in the circadian rhythm of renal functions are associated with the development of hypertension, chronic kidney disease, renal fibrosis and kidney stones. Furthermore, renal circadian clocks might interfere with the pharmacokinetics and/or pharmacodynamics of various drugs and are therefore an important consideration in the treatment of some renal diseases or disorders.
Collapse
Affiliation(s)
- Dmitri Firsov
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland.
| | - Olivier Bonny
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland. .,Service of Nephrology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland.
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide a brief summary about the current state of knowledge regarding the circadian rhythm in the regulation of normal renal function. RECENT FINDINGS There is a lack of information regarding how the circadian clock mechanisms may contribute to the development of diabetic kidney disease. We discuss recent findings regarding mechanisms that are established in diabetic kidney disease and are known to be linked to the circadian clock as possible connections between these two areas. Here, we hypothesize various mechanisms that may provide a link between the clock mechanism and kidney disease in diabetes based on available data from humans and rodent models.
Collapse
Affiliation(s)
- Olanrewaju A Olaoye
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, 1600 SW Archer Road, Box 100224, Gainesville, FL, 32610, USA
| | - Sarah H Masten
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, 1600 SW Archer Road, Box 100224, Gainesville, FL, 32610, USA
| | - Rajesh Mohandas
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, 1600 SW Archer Road, Box 100224, Gainesville, FL, 32610, USA
- North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - Michelle L Gumz
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, 1600 SW Archer Road, Box 100224, Gainesville, FL, 32610, USA.
- North Florida/South Georgia Veterans Health System, Gainesville, FL, USA.
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
35
|
Li XC, Zheng X, Chen X, Zhao C, Zhu D, Zhang J, Zhuo JL. Genetic and genomic evidence for an important role of the Na +/H + exchanger 3 in blood pressure regulation and angiotensin II-induced hypertension. Physiol Genomics 2019; 51:97-108. [PMID: 30849009 PMCID: PMC6485378 DOI: 10.1152/physiolgenomics.00122.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The sodium (Na+)/hydrogen (H+) exchanger 3 (NHE3) and sodium-potassium adenosine triphosphatase (Na+/K+-ATPase) are two of the most important Na+ transporters in the proximal tubules of the kidney. On the apical membrane side, NHE3 primarily mediates the entry of Na+ into and the exit of H+ from the proximal tubules, directly and indirectly being responsible for reabsorbing ~50% of filtered Na+ in the proximal tubules of the kidney. On the basolateral membrane side, Na+/K+-ATPase serves as a powerful engine driving Na+ out of, while pumping K+ into the proximal tubules against their concentration gradients. While the roles of NHE3 and Na+/K+-ATPase in proximal tubular Na+ transport under in vitro conditions are well recognized, their respective contributions to the basal blood pressure regulation and angiotensin II (ANG II)-induced hypertension remain poorly understood. Recently, we have been fortunate to be able to use genetically modified mouse models with global, kidney- or proximal tubule-specific deletion of NHE3 to directly determine the cause and effect relationship between NHE3, basal blood pressure homeostasis, and ANG II-induced hypertension at the whole body, kidney and/or proximal tubule levels. The purpose of this article is to review the genetic and genomic evidence for an important role of NHE3 with a focus in the regulation of basal blood pressure and ANG II-induced hypertension, as we learned from studies using global, kidney- or proximal tubule-specific NHE3 knockout mice. We hypothesize that NHE3 in the proximal tubules is necessary for maintaining basal blood pressure homeostasis and the development of ANG II-induced hypertension.
Collapse
Affiliation(s)
- Xiao C Li
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Xiaowen Zheng
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Xu Chen
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Chunling Zhao
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Dongmin Zhu
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Jianfeng Zhang
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Jia L Zhuo
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
36
|
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
|
37
|
Douma LG, Solocinski K, Holzworth MR, Crislip GR, Masten SH, Miller AH, Cheng KY, Lynch IJ, Cain BD, Wingo CS, Gumz ML. Female C57BL/6J mice lacking the circadian clock protein PER1 are protected from nondipping hypertension. Am J Physiol Regul Integr Comp Physiol 2018; 316:R50-R58. [PMID: 30427705 DOI: 10.1152/ajpregu.00381.2017] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The circadian clock is integral to the maintenance of daily rhythms of many physiological outputs, including blood pressure. Our laboratory has previously demonstrated the importance of the clock protein period 1 (PER1) in blood pressure regulation in male mice. Briefly, a high-salt diet (HS; 4% NaCl) plus injection with the long-acting mineralocorticoid deoxycorticosterone pivalate (DOCP) resulted in nondipping hypertension [<10% difference between night and day blood pressure (BP) in Per1-knockout (KO) mice but not in wild-type (WT) mice]. To date, there have been no studies that have examined the effect of a core circadian gene KO on BP rhythms in female mice. The goal of the present study was to determine whether female Per1-KO mice develop nondipping hypertension in response to HS/DOCP treatment. For the first time, we demonstrate that loss of the circadian clock protein PER1 in female mice does not significantly change mean arterial pressure (MAP) or the BP rhythm relative to female C57BL/6 WT control mice. Both WT and Per1-KO female mice experienced a significant increase in MAP in response to HS/DOCP. Importantly, however, both genotypes maintained a >10% dip in BP on HS/DOCP. This effect is distinct from the nondipping hypertension seen in male Per1-KO mice, demonstrating that the female sex appears to be protective against PER1-mediated nondipping hypertension in response to HS/DOCP. Together, these data suggest that PER1 acts in a sex-dependent manner in the regulation of cardiovascular rhythms.
Collapse
Affiliation(s)
- Lauren G Douma
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida
| | - Kristen Solocinski
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida
| | | | - G Ryan Crislip
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| | - Sarah H Masten
- Department of Medicine, University of Florida , Gainesville, Florida
| | - Amber H Miller
- Department of Medicine, University of Florida , Gainesville, Florida
| | - Kit-Yan Cheng
- Department of Medicine, University of Florida , Gainesville, Florida
| | - I Jeanette Lynch
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| | - Brian D Cain
- Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida
| | - Charles S Wingo
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida.,Research, North Florida/South Georgia Veterans Health System, Gainesville, Florida
| | - Michelle L Gumz
- Department of Medicine, 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
| |
Collapse
|
38
|
De Lavallaz L, Musso CG. Chronobiology in nephrology: the influence of circadian rhythms on renal handling of drugs and renal disease treatment. Int Urol Nephrol 2018; 50:2221-2228. [PMID: 30324579 DOI: 10.1007/s11255-018-2001-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 10/04/2018] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Chronobiology studies the phenomenon of rhythmicity in living organisms. The circadian rhythms are genetically determined and regulated by external synchronizers (the daylight cycle). Several biological processes involved in the pharmacokinetics and pharmacodynamics of drugs are subjected to circadian variations. Chronopharmacology studies how biological rhythms influence pharmacokinetics, pharmacodynamics, and toxicity, and determines whether time-of-day administration modifies the pharmacological characteristics of the drug. Chronotherapy applies chronopharmacological studies to clinical treatments, determining the best biological time for dosing: when the beneficial effects are maximal and the incidence and/or intensity of related side effects and toxicity are minimal. Most water-soluble drugs or drug metabolites are eliminated by urine through the kidney. The rate of drug clearance in the urine depends on several intrinsic variables related to renal function including renal blood flow, glomerular filtration rate, the ability of the kidney to reabsorb or to secrete drugs, urine flow, and urine pH, which influences the degree of urine acidification. Curiously, all these variables present a circadian behavior in different mammalian models. CONCLUSION The circadian rhythms have influence in the renal physiology, pathophysiology, and pharmacology, and these data should be taken into account in clinical nephrology practice.
Collapse
Affiliation(s)
- Lucas De Lavallaz
- Human Physiology Department, Instituto Universitario del Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Carlos G Musso
- Human Physiology Department, Instituto Universitario del Hospital Italiano de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
39
|
Alicic RZ, Johnson EJ, Tuttle KR. SGLT2 Inhibition for the Prevention and Treatment of Diabetic Kidney Disease: A Review. Am J Kidney Dis 2018; 72:267-277. [DOI: 10.1053/j.ajkd.2018.03.022] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/06/2018] [Indexed: 02/06/2023]
|
40
|
Crislip GR, Masten SH, Gumz ML. RECENT ADVANCES IN UNDERSTANDING THE CIRCADIAN CLOCK IN RENAL PHYSIOLOGY. CURRENT OPINION IN PHYSIOLOGY 2018; 5:38-44. [PMID: 30714020 DOI: 10.1016/j.cophys.2018.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Accumulating evidence suggests a critical role for the molecular circadian clock in the regulation of renal function. Here, we consider the most recent advances in our understanding of the relationship between the circadian clock and renal physiology.
Collapse
Affiliation(s)
- G Ryan Crislip
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation.,Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610
| | - Sarah H Masten
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation
| | - Michelle L Gumz
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation.,Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610
| |
Collapse
|
41
|
García Nieto VM, Pérez Bastida XI, Salvador Cañibano M, García Rodríguez VE, Monge Zamorano M, Luis Yanes MI. Cuantificación del riesgo de formación de cálculos cálcicos en la orina correspondiente a 2 momentos del día en un grupo de niños estudiados para descartar prelitiasis. Nefrologia 2018; 38:267-272. [DOI: 10.1016/j.nefro.2017.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 06/12/2017] [Accepted: 07/20/2017] [Indexed: 10/18/2022] Open
|
42
|
Abstract
The kidneys regulate many vital functions that require precise control throughout the day. These functions, such as maintaining sodium balance or regulating arterial pressure, rely on an intrinsic clock mechanism that was commonly believed to be controlled by the central nervous system. Mounting evidence in recent years has unveiled previously underappreciated depth of influence by circadian rhythms and clock genes on renal function, at the molecular and physiological level, independent of other external factors. The impact of circadian rhythms in the kidney also affects individuals from a clinical standpoint, as the loss of rhythmic activity or clock gene expression have been documented in various cardiovascular diseases. Fortunately, the prognostic value of examining circadian rhythms may prove useful in determining the progression of a kidney-related disease, and chronotherapy is a clinical intervention that requires consideration of circadian and diurnal rhythms in the kidney. In this review, we discuss evidence of circadian regulation in the kidney from basic and clinical research in order to provide a foundation on which a great deal of future research is needed to expand our understanding of circadian relevant biology.
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, United States
| | - David M Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
43
|
Douma LG, Gumz ML. Circadian clock-mediated regulation of blood pressure. Free Radic Biol Med 2018; 119:108-114. [PMID: 29198725 PMCID: PMC5910276 DOI: 10.1016/j.freeradbiomed.2017.11.024] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/21/2017] [Accepted: 11/28/2017] [Indexed: 12/24/2022]
Abstract
Most bodily functions vary over the course of a 24h day. Circadian rhythms in body temperature, sleep-wake cycles, metabolism, and blood pressure (BP) are just a few examples. These circadian rhythms are controlled by the central clock in the suprachiasmatic nucleus (SCN) of the hypothalamus and peripheral clocks located throughout the body. Light and food cues entrain these clocks to the time of day and this synchronicity contributes to the regulation of a variety of physiological processes with effects on overall health. The kidney, brain, nervous system, vasculature, and heart have been identified through the use of mouse models and clinical trials as peripheral clock regulators of BP. The dysregulation of this circadian pattern of BP, with or without hypertension, is associated with increased risk for cardiovascular disease. The mechanism of this dysregulation is unknown and is a growing area of research. In this review, we highlight research of human and mouse circadian models that has provided insight into the roles of these molecular clocks and their effects on physiological functions. Additional tissue-specific studies of the molecular clock mechanism are needed, as well as clinical studies including more diverse populations (different races, female patients, etc.), which will be critical to fully understand the mechanism of circadian regulation of BP. Understanding how these molecular clocks regulate the circadian rhythm of BP is critical in the treatment of circadian BP dysregulation and hypertension.
Collapse
Affiliation(s)
- Lauren G Douma
- Department of Medicine, Division of Nephrology, Hypertension and Renal Transplantation, University of Florida, Gainesville, FL 32610, United States; Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, United States
| | - Michelle L Gumz
- Department of Medicine, Division of Nephrology, Hypertension and Renal Transplantation, University of Florida, Gainesville, FL 32610, United States; Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, United States.
| |
Collapse
|
44
|
Chonlaket P, Wongwan T, Soodvilai S. Liver X receptor activation inhibits SGLT2-mediated glucose transport in human renal proximal tubular cells. Exp Physiol 2018; 103:250-260. [PMID: 29127736 DOI: 10.1113/ep086478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/10/2017] [Indexed: 12/27/2022]
Abstract
NEW FINDINGS What is the central question of this study? The liver X receptor (LXR) has been reported to regulate several membrane transporters. It is imperative to investigate whether LXR activation regulates SGLT2-mediated glucose transport in human renal proximal tubular cells. What is the main finding and its importance? Liver X receptor activation inhibits SGLT2 transport function in normal and high-glucose conditions via reduction of SGLT2 protein expression. Liver X receptors (LXRs) are members of a nuclear receptor family consisting of two isoforms, LXRα and LXRβ. They play a major role in energy metabolism, including lipid and glucose metabolism. Recent studies reported that LXRs regulate plasma glucose, although the mechanism is still uncertain. The present study investigated whether LXR activation regulates sodium glucose cotransporter2 (SGLT2) in human renal proximal tubular cells. LXR agonists, T0901317 and GW3965, inhibited SGLT2-mediated glucose uptake in a concentration-dependent manner. The effect of T0901317 and GW3965 was attenuated by a LXR antagonist, fenofibrate. Activation of the retinoid X receptor (RXR) agonist, bexarotene, potentiates the inhibitory effect of these ligands. Thus, the inhibitory effect of LXR agonists on SGLT2 was mediated and facilitated by LXR and RXR activation, respectively. In addition, the inhibitory effect of LXR agonists was not mediated by cytotoxicity. Exposing HK-2 cells, a renal proximal tubular cell line, to LXR agonists significantly reduced the maximal transport rate of SGLT2 without any effect on transporter affinity. Western blot analysis revealed that LXR activation significantly decreased protein expression of SGLT2 with no change in mRNA level. In addition, LXR activation inhibited canagliflozin-sensitive short-circuit current, which represents SGLT2-mediated glucose transport in a polarized human renal proximal tubular cell monolayer. Furthermore, LXR activation inhibited the transport function of SGLT2 in hyperglycaemic conditions. As such, this study represents evidence for the inhibitory effect of LXR activation on glucose transport in human renal proximal tubular cells.
Collapse
Affiliation(s)
- Pattira Chonlaket
- Toxicology Graduate Program, Multidisciplinary Unit, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Research Center of Transporter Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Teerasak Wongwan
- Research Center of Transporter Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Sunhapas Soodvilai
- Research Center of Transporter Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
45
|
Altered Circadian Timing System-Mediated Non-Dipping Pattern of Blood Pressure and Associated Cardiovascular Disorders in Metabolic and Kidney Diseases. Int J Mol Sci 2018; 19:ijms19020400. [PMID: 29385702 PMCID: PMC5855622 DOI: 10.3390/ijms19020400] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/12/2018] [Accepted: 01/20/2018] [Indexed: 12/15/2022] Open
Abstract
The morning surge in blood pressure (BP) coincides with increased cardiovascular (CV) events. This strongly suggests that an altered circadian rhythm of BP plays a crucial role in the development of CV disease (CVD). A disrupted circadian rhythm of BP, such as the non-dipping type of hypertension (i.e., absence of nocturnal BP decline), is frequently observed in metabolic disorders and chronic kidney disease (CKD). The circadian timing system, controlled by the central clock in the suprachiasmatic nucleus of the hypothalamus and/or by peripheral clocks in the heart, vasculature, and kidneys, modulates the 24 h oscillation of BP. However, little information is available regarding the molecular and cellular mechanisms of an altered circadian timing system-mediated disrupted dipping pattern of BP in metabolic disorders and CKD that can lead to the development of CV events. A more thorough understanding of this pathogenesis could provide novel therapeutic strategies for the management of CVD. This short review will address our and others' recent findings on the molecular mechanisms that may affect the dipping pattern of BP in metabolic dysfunction and kidney disease and its association with CV disorders.
Collapse
|
46
|
Douma LG, Holzworth MR, Solocinski K, Masten SH, Miller AH, Cheng KY, Lynch IJ, Cain BD, Wingo CS, Gumz ML. Renal Na-handling defect associated with PER1-dependent nondipping hypertension in male mice. Am J Physiol Renal Physiol 2018; 314:F1138-F1144. [PMID: 29357420 DOI: 10.1152/ajprenal.00546.2017] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Many physiological functions have a circadian rhythm, including blood pressure (BP). BP is highest during the active phase, whereas during the rest period, BP dips 10-20%. Patients that do not experience this dip at night are termed "nondippers." Nondipping hypertension is associated with increased risk of cardiovascular disease. The mechanisms underlying nondipping hypertension are not understood. Without the circadian clock gene Per1, C57BL/6J mice develop nondipping hypertension on a high-salt diet plus mineralocorticoid treatment (HS/DOCP). Our laboratory has shown that PER1 regulates expression of several genes related to sodium (Na) transport in the kidney, including epithelial Na channel (ENaC) and Na chloride cotransporter (NCC). Urinary Na excretion also demonstrates a circadian pattern with a peak during active periods. We hypothesized that PER1 contributes to circadian regulation of BP via a renal Na-handling-dependent mechanism. Na-handling genes from the distal nephron were inappropriately regulated in KO mice on HS/DOCP. Additionally, the night/day ratio of Na urinary excretion by Per1 KO mice is decreased compared with WT (4 × vs. 7×, P < 0.001, n = 6 per group). Distal nephron-specific Per1 KO mice also show an inappropriate increase in expression of Na transporter genes αENaC and NCC. These results support the hypothesis that PER1 mediates control of circadian BP rhythms via the regulation of distal nephron Na transport genes. These findings have implications for the understanding of the etiology of nondipping hypertension and the subsequent development of novel therapies for this dangerous pathophysiological condition.
Collapse
Affiliation(s)
- Lauren G Douma
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida
| | | | - Kristen Solocinski
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida
| | - Sarah H Masten
- Department of Medicine, University of Florida , Gainesville, Florida
| | - Amber H Miller
- Department of Medicine, University of Florida , Gainesville, Florida
| | - Kit-Yan Cheng
- Department of Medicine, University of Florida , Gainesville, Florida
| | - I Jeanette Lynch
- Department of Medicine, University of Florida , Gainesville, Florida.,Research, North Florida/South Georgia Veterans Health System, Gainesville, Florida
| | - Brian D Cain
- Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida
| | - Charles S Wingo
- Department of Medicine, University of Florida , Gainesville, Florida.,Research, North Florida/South Georgia Veterans Health System, Gainesville, Florida
| | - Michelle L Gumz
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida.,Research, North Florida/South Georgia Veterans Health System, Gainesville, Florida
| |
Collapse
|
47
|
Gumz ML. Molecular basis of circadian rhythmicity in renal physiology and pathophysiology. Exp Physiol 2018; 101:1025-9. [PMID: 27474264 DOI: 10.1113/ep085781] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/20/2016] [Indexed: 12/20/2022]
Abstract
NEW FINDINGS What is the topic of this review? This brief symposium report is focused on the molecular and physiological evidence that supports a key role for the circadian clock in the regulation of kidney function. What advances does it highlight? Progress in understanding the molecular mechanism of the kidney clock is reviewed here, including new results from global 'omics' studies and candidate gene approaches. The molecular kidney clock is a master regulator of gene expression that affects renal electrolyte and drug handling as well as blood pressure. In this brief review, an overview of the molecular and physiological evidence for the kidney clock and the implications for the regulation of renal physiology and pathophysiology are presented. Accumulating evidence suggests that the molecular circadian clock acts as a master regulator of gene expression in the kidney. Global transcriptomic approaches have revealed the important finding that there are thousands of genes in the kidney subject to regulation by the molecular clock. Candidate gene approaches have also yielded information regarding regulation of renal sodium transport genes by the molecular clock. To date, the evidence linking the molecular kidney clock to rhythmic renal function provides strong support for the concept that circadian control of gene expression underlies rhythms in physiological function.
Collapse
Affiliation(s)
- Michelle L Gumz
- Department of Medicine, Division of Nephrology, Hypertension and Renal Transplantation, University of Florida, Gainesville, FL, USA.,Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
48
|
Solocinski K, Holzworth M, Wen X, Cheng KY, Lynch IJ, Cain BD, Wingo CS, Gumz ML. Desoxycorticosterone pivalate-salt treatment leads to non-dipping hypertension in Per1 knockout mice. Acta Physiol (Oxf) 2017; 220:72-82. [PMID: 27636900 DOI: 10.1111/apha.12804] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 08/06/2016] [Accepted: 09/11/2016] [Indexed: 12/01/2022]
Abstract
AIM Increasing evidence demonstrates that circadian clock proteins are important regulators of physiological functions including blood pressure. An established risk factor for developing cardiovascular disease is the absence of a blood pressure dip during the inactive period. The goal of the present study was to determine the effects of a high salt diet plus mineralocorticoid on PER1-mediated blood pressure regulation in a salt-resistant, normotensive mouse model, C57BL/6J. METHODS Blood pressure was measured using radiotelemetry. After control diet, wild-type (WT) and Per1 (KO) knockout mice were given a high salt diet (4% NaCl) and the long-acting mineralocorticoid deoxycorticosterone pivalate. Blood pressure and activity rhythms were analysed to evaluate changes over time. RESULTS Blood pressure in WT mice was not affected by a high salt diet plus mineralocorticoid. In contrast, Per1 KO mice exhibited significantly increased mean arterial pressure (MAP) in response to a high salt diet plus mineralocorticoid. The inactive/active phase ratio of MAP in WT mice was unchanged by high salt plus mineralocorticoid treatment. Importantly, this treatment caused Per1 KO mice to lose the expected decrease or 'dip' in blood pressure during the inactive compared to the active phase. CONCLUSION Loss of PER1 increased sensitivity to the high salt plus mineralocorticoid treatment. It also resulted in a non-dipper phenotype in this model of salt-sensitive hypertension and provides a unique model of non-dipping. Together, these data support an important role for the circadian clock protein PER1 in the modulation of blood pressure in a high salt/mineralocorticoid model of hypertension.
Collapse
Affiliation(s)
- K Solocinski
- Department of Medicine, University of Florida, Gainesville, FL, USA
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - M Holzworth
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - X Wen
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - K-Y Cheng
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - I J Lynch
- Department of Medicine, University of Florida, Gainesville, FL, USA
- North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - B D Cain
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - C S Wingo
- Department of Medicine, University of Florida, Gainesville, FL, USA
- North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - M L Gumz
- Department of Medicine, University of Florida, Gainesville, FL, USA
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
- North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| |
Collapse
|
49
|
Inhibition of expression of the circadian clock gene Period causes metabolic abnormalities including repression of glycometabolism in Bombyx mori cells. Sci Rep 2017; 7:46258. [PMID: 28393918 PMCID: PMC5385517 DOI: 10.1038/srep46258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/13/2017] [Indexed: 02/07/2023] Open
Abstract
Abnormalities in the circadian clock system are known to affect the body’s metabolic functions, though the molecular mechanisms responsible remain uncertain. In this study, we achieved continuous knockdown of B. mori Period (BmPer) gene expression in the B. mori ovary cell line (BmN), and generated a Per-KD B. mori model with developmental disorders including small individual cells and slow growth. We conducted cell metabolomics assays by gas chromatography/liquid chromatography-mass spectrometry and showed that knockdown of BmPer gene expression resulted in significant inhibition of glycometabolism. Amino acids that used glucose metabolites as a source were also down-regulated, while lipid metabolism and nucleotide metabolism were significantly up-regulated. Metabolite correlation analysis showed that pyruvate and lactate were closely related to glycometabolism, as well as to metabolites such as aspartate, alanine, and xanthine in other pathways. Further validation experiments showed that the activities of the key enzymes of glucose metabolism, hexokinase, phosphofructokinase, and citrate synthase, were significantly decreased and transcription of their encoding genes, as well as that of pyruvate kinase, were also significantly down-regulated. We concluded that inhibition of the circadian clock gene BmPer repressed glycometabolism, and may be associated with changes in cellular amino acid metabolism, and in cell growth and development.
Collapse
|
50
|
Dominguez Rieg JA, de la Mora Chavez S, Rieg T. Novel developments in differentiating the role of renal and intestinal sodium hydrogen exchanger 3. Am J Physiol Regul Integr Comp Physiol 2016; 311:R1186-R1191. [PMID: 27733387 DOI: 10.1152/ajpregu.00372.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 09/22/2016] [Accepted: 10/05/2016] [Indexed: 11/22/2022]
Abstract
The Na+/H+ exchanger isoform 3 (NHE3) facilitates Na+ absorption and H+ secretion and is expressed in the intestine, proximal tubule, and thick ascending limb of the kidney. While the function of NHE3 for Na+ and [Formula: see text](re)absorption has been defined using conventional NHE3 knockout mice (NHE3-/-), the recent generation of conditional NHE3 knockout mice started to give critical new insight into the role of this protein by allowing for temporal and spatial control of NHE3 expression. For example, in contrast to NHE3-/- mice, knockout of NHE3 in the S1 and S2 segments of the proximal tubule or along the entire tubule/collecting duct does not cause any lethality. Nonabsorbable NHE3 inhibitors have been developed, and preclinical as well as clinical trials indicate possible pharmacological use in fluid overload, hypertension, chronic kidney disease, hyperphosphatemia, and constipation. Some of the therapeutic considerations seem to be directly related to the pharmacodynamic properties of these drugs; however, little is known about the effects of these nonabsorbable NHE3 inhibitors on intestinal phosphate transport and the mechanisms so far remain elusive. This review focuses on novel findings of NHE3 in the intestine and the kidney as well as novel drug developments targeting NHE3.
Collapse
Affiliation(s)
- Jessica A Dominguez Rieg
- Department of Basic Sciences, Bastyr University California, San Diego, California.,Veterans Affairs San Diego Healthcare System, San Diego, California; and
| | | | - Timo Rieg
- Veterans Affairs San Diego Healthcare System, San Diego, California; and .,Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|