1
|
Stadt M, Layton AT. Modulation of blood pressure by dietary potassium and sodium: sex differences and modeling analysis. Am J Physiol Renal Physiol 2025; 328:F406-F417. [PMID: 39447116 DOI: 10.1152/ajprenal.00222.2024] [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: 07/30/2024] [Revised: 09/05/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
High Na+ intake has been linked to elevations in blood pressure, whereas K+ has the opposite effect. The underlying mechanisms involve complex interactions among renal function, fluid volume, fluid-regulatory hormones, vasculature, cardiac function, and the autonomic nervous system. These mechanisms are likely modulated by sex, given the known sex differences in blood pressure regulation and the higher prevalence of hypertension in men. The source of these observed sex differences may be traced to organ and tissue levels, given that kidney function, intrarenal renin-angiotensin system components, renal sympathetic nervous activity, and nitric oxide bioavailability all exhibit sex differences. To assess the functional impact of each of these sex differences, we developed sex-specific computational models to simulate whole-body Na+, K+, and fluid homeostasis, and the effects on blood pressure. The models describe the interactions among the renal system, cardiovascular system, gastrointestinal system, renal sympathetic nervous system, and renin-angiotensin-aldosterone system. Model simulations suggest that women's attenuated blood pressure response to hypertensive stimuli, including high Na+ intake, may be largely attributable to the female renal transporter abundance pattern. In addition, we investigated the causal link between high K+ intake and blood pressure reduction. The models simulate renal response to high K+ intake, including the immediate gastrointestinal feedforward signals to the kidneys to increase K+ excretion, and the longer-term response to decrease proximal fractional Na+ reabsorption and distal K+ reabsorption. With these assumptions, simulations of high K+ intake yielded kaliuresis, natriuresis, and a substantial reduction in blood pressure, even when combined with high Na+ intake.NEW & NOTEWORTHY Excessive dietary Na+ raises blood pressure, whereas a high K+ diet has the opposite effect. The underlying mechanisms are moderated by sex and involve multiple organs and tissues. How do high K+-induced alternations in kidney function lower blood pressure, and how do those mechanisms differ between men and women? To answer these questions, we conducted computer simulations to simulate whole-body fluid and electrolyte homeostasis, and the effects of Na+ and K+ intake on blood pressure.
Collapse
Affiliation(s)
- Melissa Stadt
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Anita T Layton
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
- Cheriton School of Computer Science, University of Waterloo, Waterloo, Ontario, Canada
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
2
|
Masenga SK, Wandira N, Cattivelli-Murdoch G, Saleem M, Beasley H, Hinton A, Ertuglu LA, Mwesigwa N, Kleyman TR, Kirabo A. Salt sensitivity of blood pressure: mechanisms and sex-specific differences. Nat Rev Cardiol 2025:10.1038/s41569-025-01135-0. [PMID: 39984695 DOI: 10.1038/s41569-025-01135-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 02/23/2025]
Abstract
Salt sensitivity of blood pressure (SSBP) is an independent risk factor for cardiovascular disease in individuals with or without hypertension. However, the mechanisms and management of SSBP remain unclear, mainly because the diagnosis of this condition relies on salt loading-depletion protocols that are not feasible in the clinic. The prevalence of hypertension is lower in premenopausal women than in men, but this sex-specific difference is reversed after menopause. Whether excessive SSBP in women at any age contributes to this reversal is unknown, but many clinical studies that have rigorously assessed for SSBP using salt loading-depletion protocols have confirmed that SSBP is more prevalent in women than in men, including during premenopausal age. In this Review, we discuss sex-specific mechanisms of SSBP. We describe sex-related differences in renal transporters, hypertensive pregnancy, SSBP in autoimmune disorders and mitogen-activated protein kinase signalling pathways, and highlight limitations and lessons learned from Dahl salt-sensitive rat models.
Collapse
Affiliation(s)
- Sepiso K Masenga
- HAND research Group, Department of Pathology and Physiological Sciences, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia.
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Nelson Wandira
- Vanderbilt Mater of Public Health Program, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Mohammad Saleem
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heather Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Lale A Ertuglu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Naome Mwesigwa
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, Department of Cell Biology, and Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Annet Kirabo
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
3
|
Zhang H, Singal PK, Ravandi A, Rabinovich-Nikitin I. Sex-Specific Differences in the Pathophysiology of Hypertension. Biomolecules 2025; 15:143. [PMID: 39858537 PMCID: PMC11763887 DOI: 10.3390/biom15010143] [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: 11/28/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Hypertension is one of the most common comorbidities in cardiometabolic diseases, affecting nearly one third of adults. As a result, its pathophysiological mechanisms have been studied extensively and are focused around pressure natriuresis, the renin-angiotensin system (RAS), the sympathetic nervous system, oxidative stress, and endothelial dysfunction. Additionally, hypertension secondary to other underlying etiologies also exists. While clinical evidence has clearly shown differences in hypertension development in males and females, relatively little is known about the pathophysiological mechanisms behind these differences. Sex hormones likely play a key role, as they modulate many factors related to hypertension development. In this review, we postulate the potential role for sexually dimorphic fat metabolism in the physiology of hypertension. In brief, estrogen promotes subcutaneous fat deposition over visceral fat and increases in mass via adaptive hyperplasia rather than pathogenic hypertrophy. This adipose tissue subsequently produces anti-inflammatory effects and inhibits metabolic dysfunction-associated fatty liver disease (MAFLD) and RAS activation, ultimately leading to decreased levels of hypertension in pre-menopausal females. On the other hand, androgens and the lack of estrogens promote visceral and ectopic fat deposition, including in the liver, and lead to increased circulating pro-inflammatory cytokines and potentially subsequent RAS activation and hypertension development in males and post-menopausal females. Understanding the sex-specific differences in fat metabolism may provide deeper insights into the patho-mechanisms associated with hypertension and lead to more comprehensive sex-specific care.
Collapse
Affiliation(s)
- Hannah Zhang
- Department of Physiology and Pathophysiology, St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Rm. 3042, 351 Taché Avenue, Winnipeg, MB R2H 2A6, Canada; (H.Z.); (P.K.S.); (A.R.)
- Department of Pharmacology and Therapeutics, St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Rm. 3042, 351 Taché Avenue, Winnipeg, MB R2H 2A6, Canada
| | - Pawan K. Singal
- Department of Physiology and Pathophysiology, St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Rm. 3042, 351 Taché Avenue, Winnipeg, MB R2H 2A6, Canada; (H.Z.); (P.K.S.); (A.R.)
- Department of Pharmacology and Therapeutics, St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Rm. 3042, 351 Taché Avenue, Winnipeg, MB R2H 2A6, Canada
| | - Amir Ravandi
- Department of Physiology and Pathophysiology, St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Rm. 3042, 351 Taché Avenue, Winnipeg, MB R2H 2A6, Canada; (H.Z.); (P.K.S.); (A.R.)
- Department of Pharmacology and Therapeutics, St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Rm. 3042, 351 Taché Avenue, Winnipeg, MB R2H 2A6, Canada
- Section of Cardiology, Department of Medicine, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Inna Rabinovich-Nikitin
- Department of Physiology and Pathophysiology, St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Rm. 3042, 351 Taché Avenue, Winnipeg, MB R2H 2A6, Canada; (H.Z.); (P.K.S.); (A.R.)
- Department of Pharmacology and Therapeutics, St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Rm. 3042, 351 Taché Avenue, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
4
|
Kumar P, Neelamegam K, Ramasamy C, Samivel R, Xia H, Kapusta DR, Pandey KN. Epigenetic mechanisms differentially regulate blood pressure and renal dysfunction in male and female Npr1 haplotype mice. FASEB J 2024; 38:e23858. [PMID: 39109516 PMCID: PMC11309581 DOI: 10.1096/fj.202400714r] [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: 03/31/2024] [Revised: 07/02/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024]
Abstract
We determined the epigenetic mechanisms regulating mean arterial pressure (MAP) and renal dysfunction in guanylyl cyclase/natriuretic peptide receptor-A (GC-A/NPRA) gene-targeted mice. The Npr1 (encoding NPRA) gene-targeted mice were treated with class 1 specific histone deacetylase inhibitor (HDACi) mocetinostat (MGCD) to determine the epigenetic changes in a sex-specific manner. Adult male and female Npr1 haplotype (1-copy; Npr1+/-), wild-type (2-copy; Npr1+/+), and gene-duplicated heterozygous (3-copy; Npr1++/+) mice were intraperitoneally injected with MGCD (2 mg/kg) for 14 days. BP, renal function, histopathology, and epigenetic changes were measured. One-copy male mice showed significantly increased MAP, renal dysfunction, and fibrosis than 2-copy and 3-copy mice. Furthermore, HDAC1/2, collagen1alpha-2 (Col1α-2), and alpha smooth muscle actin (α-SMA) were significantly increased in 1-copy mice compared with 2-copy controls. The expression of antifibrotic microRNA-133a was attenuated in 1-copy mice but to a greater extent in males than females. NF-κB was localized at significantly lower levels in cytoplasm than in the nucleus with stronger DNA binding activity in 1-copy mice. MGCD significantly lowered BP, improved creatinine clearance, and repaired renal histopathology. The inhibition of class I HDACs led to a sex-dependent distinctive stimulation of acetylated positive histone marks and inhibition of methylated repressive histone marks in Npr1 1-copy mice; however, it epigenetically lowered MAP, repaired renal fibrosis, and proteinuria and suppressed NF-kB differentially in males versus females. Our results suggest a role for epigenetic targets affecting hypertension and renal dysfunction in a sex-specific manner.
Collapse
Affiliation(s)
- Prerna Kumar
- Department of PhysiologySchool of Medicine, Tulane University Health Sciences CenterNew OrleansLouisianaUSA
| | - Kandasamy Neelamegam
- Department of PhysiologySchool of Medicine, Tulane University Health Sciences CenterNew OrleansLouisianaUSA
| | - Chandramohan Ramasamy
- Department of PhysiologySchool of Medicine, Tulane University Health Sciences CenterNew OrleansLouisianaUSA
| | - Ramachandran Samivel
- Department of PhysiologySchool of Medicine, Tulane University Health Sciences CenterNew OrleansLouisianaUSA
| | - Huijing Xia
- Department of PharmacologyLouisiana State University Health Sciences CenterNew OrleansLouisianaUSA
| | - Daniel R. Kapusta
- Department of PharmacologyLouisiana State University Health Sciences CenterNew OrleansLouisianaUSA
| | - Kailash N. Pandey
- Department of PhysiologySchool of Medicine, Tulane University Health Sciences CenterNew OrleansLouisianaUSA
| |
Collapse
|
5
|
Turner CG, de Oliveira K, Lu Q, Patel AR, Pulakat L, Jaffe IZ, DuPont JJ. Microvascular angiotensin II type 2 receptor function is enhanced in young females and declines in a model of murine aging. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:19. [PMID: 40290368 PMCID: PMC12030185 DOI: 10.20517/jca.2024.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Introduction Angiotensin II (AngII) affects cardiovascular health, mediating impacts through AngII type 1 (AT1R) and type 2 (AT2R) receptors. The present study investigated sex and aging-related differences in microvascular AngII receptor function in mice and humans. Methods Mesenteric resistance arteries (MRA) were isolated from 3-, 12-, and 18-month-old female and male C57/Bl6 mice. Wire myography was used to measure vasoconstriction to AngII and vasodilation to an AT2R agonist (compound 21, C21). Seven healthy adults (3 premenopausal women and 4 age-matched men) were recruited to participate in a study measuring cutaneous microvascular vasoconstriction to AngII in the presence and absence of 10 μM PD123319, an AT2R antagonist. Results In murine MRA, AngII-induced constriction increases by 18 months in females and by 12 months in males. AT2R-mediated vasodilation was reduced with age in females only, which corresponds with a female-specific decrease in mesenteric AT2R mRNA expression. AT2R inhibition enhances AngII-induced constriction in young female, but not male, mice. Clinical data support that premenopausal women have attenuated AngII constriction vs. men, which is abrogated by AT2R inhibition. AT2R expression is greater in primary aortic smooth muscle cells, but not endothelial cells, from young women compared with men. Conclusions These data demonstrate enhanced microvascular AT2R function in young female mice and young women. There is a female-specific loss of AT2R function with age in mice, concomitant with declining AT2R expression. These findings implicate AT2R as a sex-specific target for microvascular dysfunction and aging-associated cardiovascular disease.
Collapse
Affiliation(s)
- Casey G. Turner
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Karla de Oliveira
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Ayan R. Patel
- Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Lakshmi Pulakat
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Jennifer J. DuPont
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| |
Collapse
|
6
|
Drury ER, Wu J, Gigliotti JC, Le TH. Sex differences in blood pressure regulation and hypertension: renal, hemodynamic, and hormonal mechanisms. Physiol Rev 2024; 104:199-251. [PMID: 37477622 PMCID: PMC11281816 DOI: 10.1152/physrev.00041.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/06/2023] [Accepted: 07/16/2023] [Indexed: 07/22/2023] Open
Abstract
The teleology of sex differences has been argued since at least as early as Aristotle's controversial Generation of Animals more than 300 years BC, which reflects the sex bias of the time to contemporary readers. Although the question "why are the sexes different" remains a topic of debate in the present day in metaphysics, the recent emphasis on sex comparison in research studies has led to the question "how are the sexes different" being addressed in health science through numerous observational studies in both health and disease susceptibility, including blood pressure regulation and hypertension. These efforts have resulted in better understanding of differences in males and females at the molecular level that partially explain their differences in vascular function and renal sodium handling and hence blood pressure and the consequential cardiovascular and kidney disease risks in hypertension. This review focuses on clinical studies comparing differences between men and women in blood pressure over the life span and response to dietary sodium and highlights experimental models investigating sexual dimorphism in the renin-angiotensin-aldosterone, vascular, sympathetic nervous, and immune systems, endothelin, the major renal sodium transporters/exchangers/channels, and the impact of sex hormones on these systems in blood pressure homeostasis. Understanding the mechanisms governing sex differences in blood pressure regulation could guide novel therapeutic approaches in a sex-specific manner to lower cardiovascular risks in hypertension and advance personalized medicine.
Collapse
Affiliation(s)
- Erika R Drury
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - Jing Wu
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, United States
| | - Joseph C Gigliotti
- Department of Integrative Physiology and Pharmacology, Liberty University College of Osteopathic Medicine, Lynchburg, Virginia, United States
| | - Thu H Le
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States
| |
Collapse
|
7
|
Mei X, Mell B, Aryal S, Manandhar I, Tummala R, Zubcevic J, Lai K, Yang T, Li Q, Yeoh BS, Joe B. Genetically engineered Lactobacillus paracasei rescues colonic angiotensin converting enzyme 2 (ACE2) and attenuates hypertension in female Ace2 knock out rats. Pharmacol Res 2023; 196:106920. [PMID: 37716548 PMCID: PMC10976180 DOI: 10.1016/j.phrs.2023.106920] [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: 05/29/2023] [Revised: 08/24/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
Engineered gut microbiota represents a new frontier in medicine, in part serving as a vehicle for the delivery of therapeutic biologics to treat a range of host conditions. The gut microbiota plays a significant role in blood pressure regulation; thus, manipulation of gut microbiota is a promising avenue for hypertension treatment. In this study, we tested the potential of Lactobacillus paracasei, genetically engineered to produce and deliver human angiotensin converting enzyme 2 (Lacto-hACE2), to regulate blood pressure in a rat model of hypertension with genetic ablation of endogenous Ace2 (Ace2-/- and Ace2-/y). Our findings reveal a sex-specific reduction in blood pressure in female (Ace2-/-) but not male (Ace2-/y) rats following colonization with the Lacto-hACE2. This beneficial effect of lowering blood pressure was aligned with a specific reduction in colonic angiotensin II, but not renal angiotensin II, suggesting the importance of colonic Ace2 in the regulation of blood pressure. We conclude that this approach of targeting the colon with engineered bacteria for delivery of ACE2 represents a promising new paradigm in the development of antihypertensive therapeutics.
Collapse
Affiliation(s)
- Xue Mei
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Blair Mell
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Sachin Aryal
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Ishan Manandhar
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Ramakumar Tummala
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Jasenka Zubcevic
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Khanh Lai
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Tao Yang
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Qiuhong Li
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Beng San Yeoh
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
8
|
Ulpiano Trillig A, Damianaki A, Hendriks-Balk M, Brito W, Garessus J, Burnier M, Wuerzner G, Pruijm M. Determinants of Renal Micro-Perfusion as Assessed with Contrast-Enhanced Ultrasound in Healthy Males and Females. J Clin Med 2023; 12:4141. [PMID: 37373834 DOI: 10.3390/jcm12124141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
(1) Background: The renal microcirculation is essential to maintain the renal function, but its determinants in humans have been poorly studied. Contrast-enhanced ultrasound (CEUS) allows the non-invasive quantification of the cortical micro-perfusion at the bedside using the perfusion index (PI). The aims of this study were to assess whether differences exist in PI between healthy males and females and to identify clinical determinants associated with cortical micro-perfusion. (2) Methods: Healthy, normotensive volunteers (eGFR > 60 mL/min/1.73 m2, no albuminuria) underwent CEUS under standardized conditions with the destruction-reperfusion (DR) technique. The mean PI of four DR sequences was reported as the primary outcome measure (3) Results: A total of 115 subjects (77 females and 38 males) completed the study; the mean ± SD age was, respectively, 37.1 ± 12.2 and 37.1 ± 12.7 years in females and males, and the mean eGFR was 105.9 ± 15.1 and 91.0 ± 17.4 mL/min/1.73 m2. The PI (median) was higher in females than in males, i.e., 2705 (IQR 1641-3777) vs. 1965 (IQR 1294-3346) arbitrary units (a.u), p = 0.02). A correlation analysis showed positive associations between PI and eGFR, female sex, heart rate, plasma renin activity (PRA) and plasma aldosterone concentrations (PAC), negative associations with potassium, bicarbonate and systolic blood pressure, and no associations with age, body mass index and renal resistive index (RRI). In a multivariate linear regression analysis, only PRA remained significantly associated with PI. (4) Conclusions: Although the PI was higher among females, this association was no longer significant after adjustment for covariates. There was no difference in females tested during the follicular or the luteal phases. In conclusion, the PI was only weakly influenced by classic clinical variables, but was positively associated with PRA, suggesting that the renin-angiotensin system plays a role in the regulation of the cortical micro-perfusion in humans. Identifying which other factors contribute to the large variations in micro-perfusion across individuals needs further study.
Collapse
Affiliation(s)
- Antonio Ulpiano Trillig
- Service of Nephrology and Hypertension, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 17, 1005 Lausanne, Switzerland
| | - Aikaterini Damianaki
- Service of Nephrology and Hypertension, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 17, 1005 Lausanne, Switzerland
| | - Mariëlle Hendriks-Balk
- Service of Nephrology and Hypertension, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 17, 1005 Lausanne, Switzerland
| | - Wendy Brito
- Service of Nephrology and Hypertension, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 17, 1005 Lausanne, Switzerland
| | - Jonas Garessus
- Service of Nephrology and Hypertension, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 17, 1005 Lausanne, Switzerland
| | - Michel Burnier
- Service of Nephrology and Hypertension, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 17, 1005 Lausanne, Switzerland
| | - Grégoire Wuerzner
- Service of Nephrology and Hypertension, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 17, 1005 Lausanne, Switzerland
| | - Menno Pruijm
- Service of Nephrology and Hypertension, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 17, 1005 Lausanne, Switzerland
| |
Collapse
|
9
|
Chambers LC, Yen M, Jackson WF, Dorrance AM. Female mice are protected from impaired parenchymal arteriolar TRPV4 function and impaired cognition in hypertension. Am J Physiol Heart Circ Physiol 2023; 324:H581-H597. [PMID: 36897751 PMCID: PMC10069981 DOI: 10.1152/ajpheart.00481.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/11/2023]
Abstract
Hypertension is a leading modifiable risk factor for cerebral small vessel disease. Our laboratory has shown that endothelium-dependent dilation in cerebral parenchymal arterioles (PAs) is dependent on transient receptor potential vanilloid 4 (TRPV4) activation, and this pathway is impaired in hypertension. This impaired dilation is associated with cognitive deficits and neuroinflammation. Epidemiological evidence suggests that women with midlife hypertension have an increased dementia risk that does not exist in age-matched men, though the mechanisms responsible for this are unclear. This study aimed to determine the sex differences in young, hypertensive mice to serve as a foundation for future determination of sex differences at midlife. We tested the hypothesis that young hypertensive female mice would be protected from the impaired TRPV4-mediated PA dilation and cognitive dysfunction observed in male mice. Angiotensin II (ANG II)-filled osmotic minipumps (800 ng/kg/min, 4 wk) were implanted in 16- to 19-wk-old male C56BL/6 mice. Age-matched female mice received either 800 ng/kg/min or 1,200 ng/kg/min ANG II. Sham-operated mice served as controls. Systolic blood pressure was elevated in ANG II-treated male mice and in 1,200 ng ANG II-treated female mice versus sex-matched shams. PA dilation in response to the TRPV4 agonist GSK1016790A (10-9-10-5 M) was impaired in hypertensive male mice, which was associated with cognitive dysfunction and neuroinflammation, reproducing our previous findings. Hypertensive female mice exhibited normal TRPV4-mediated PA dilation and were cognitively intact. Female mice also showed fewer signs of neuroinflammation than male mice. Determining the sex differences in cerebrovascular health in hypertension is critical for developing effective therapeutic strategies for women.NEW & NOTEWORTHY Vascular dementia is a significant public health concern, and the effect of biological sex on dementia development is not well understood. TRPV4 channels are essential regulators of cerebral parenchymal arteriolar function and cognition. Hypertension impairs TRPV4-mediated dilation and memory in male rodents. Data presented here suggest female sex protects against impaired TRPV4 dilation and cognitive dysfunction during hypertension. These data advance our understanding of the influence of biological sex on cerebrovascular health in hypertension.
Collapse
Affiliation(s)
- Laura C Chambers
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States
| | - Martina Yen
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States
| | - William F Jackson
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, Michigan, United States
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, United States
| |
Collapse
|
10
|
Biwer LA, Lu Q, Ibarrola J, Stepanian A, Man JJ, Carvajal BV, Camarda ND, Zsengeller Z, Skurnik G, Seely EW, Karumanchi SA, Jaffe IZ. Smooth Muscle Mineralocorticoid Receptor Promotes Hypertension After Preeclampsia. Circ Res 2023; 132:674-689. [PMID: 36815487 PMCID: PMC10119809 DOI: 10.1161/circresaha.122.321228] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND Preeclampsia is a syndrome of high blood pressure (BP) with end organ damage in late pregnancy that is associated with high circulating soluble VEGF receptor (sFlt1 [soluble Fms-like tyrosine kinase 1]). Women exposed to preeclampsia have a substantially increased risk of hypertension after pregnancy, but the mechanism remains unknown, leaving a missed interventional opportunity. After preeclampsia, women have enhanced sensitivity to hypertensive stress. Since smooth muscle cell mineralocorticoid receptors (SMC-MR) are activated by hypertensive stimuli, we hypothesized that high sFlt1 exposure in pregnancy induces a postpartum state of enhanced SMC-MR responsiveness. METHODS Postpartum BP response to high salt intake was studied in women with prior preeclampsia. MR transcriptional activity was assessed in vitro in sFlt1-treated SMC by reporter assays and PCR. Preeclampsia was modeled by transient sFlt1 expression in pregnant mice. Two months post-partum, mice were exposed to high salt and then to AngII (angiotensin II) and BP and vasoconstriction were measured. RESULTS Women exposed to preeclampsia had significantly enhanced salt sensitivity of BP verses those with a normotensive pregnancy. sFlt1 overexpression during pregnancy in mice induced elevated BP and glomerular endotheliosis, which resolved post-partum. The sFlt1 exposed post-partum mice had significantly increased BP response to 4% salt diet and to AngII infusion. In vitro, SMC-MR transcriptional activity in response to aldosterone or AngII was significantly increased after transient exposure to sFlt1 as was aldosterone-induced expression of AngII type 1 receptor. Post-partum, SMC-MR-KO mice were protected from the enhanced response to hypertensive stimuli after preeclampsia. Mechanistically, preeclampsia mice exposed to postpartum hypertensive stimuli develop enhanced aortic stiffness, microvascular myogenic tone, AngII constriction, and AngII type 1 receptor expression, all of which were prevented in SMC-MR-KO littermates. CONCLUSIONS These data support that sFlt1-induced vascular injury during preeclampsia produces a persistent state of enhanced sensitivity of SMC-MR to activation. This contributes to postpartum hypertension in response to common stresses and supports testing of MR antagonism to mitigate the increased cardiovascular risk in women after PE.
Collapse
Affiliation(s)
- Lauren A. Biwer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
| | - Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
| | - Alec Stepanian
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA
| | - Joshua J. Man
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA
| | - Brigett V. Carvajal
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA
| | - Nicholas D. Camarda
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA
| | | | | | - Ellen W. Seely
- Division of Endocrinology, Brigham and Women’s Hospital, Boston MA
| | - S. Ananth Karumanchi
- Department of Medicine, Beth Israel Deaconess Hospital, Boston MA
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles CA
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA
| |
Collapse
|
11
|
Abstract
Several clinical and large population studies indicate that women are more salt-sensitive than men, yet the precise mechanisms by which the sexually dimorphic onset manifests remains incompletely understood. Here, we evaluate recent epidemiological data and highlight current knowledge from studies investigating sex-specific mechanisms of salt-sensitive blood pressure (SSBP). Emerging evidence indicates that women of all ethnicities are more salt-sensitive than men, at all ages both premenopausal and postmenopausal. However, menopause exacerbates severity and prevalence of SSBP, suggesting that female sex chromosomes predispose to and female sex hormones mitigate SSBP. Results from both human and rodent studies support the contribution of enhanced and inappropriate activation of the aldosterone-ECMR (endothelial cell mineralocorticoid receptor) axis promoting vascular dysfunction in females. Increases in adrenal response to angiotensin II, in association with higher ECMR expression and activation of endothelial ENaC (epithelial sodium channel) in females compared to males, are emerging as central players in the development of endothelial dysfunction and SSBP in females. Female sex increases the prevalence and susceptibility of SSBP and sex hormones and sex chromosome complement may exert antagonistic effects in the development of the female heightened SSBP.
Collapse
Affiliation(s)
- Candee T. Barris
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jessica L. Faulkner
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Physiology Department, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Eric J. Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
12
|
Steckelings UM, Widdop RE, Sturrock ED, Lubbe L, Hussain T, Kaschina E, Unger T, Hallberg A, Carey RM, Sumners C. The Angiotensin AT 2 Receptor: From a Binding Site to a Novel Therapeutic Target. Pharmacol Rev 2022; 74:1051-1135. [PMID: 36180112 PMCID: PMC9553111 DOI: 10.1124/pharmrev.120.000281] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/19/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
Discovered more than 30 years ago, the angiotensin AT2 receptor (AT2R) has evolved from a binding site with unknown function to a firmly established major effector within the protective arm of the renin-angiotensin system (RAS) and a target for new drugs in development. The AT2R represents an endogenous protective mechanism that can be manipulated in the majority of preclinical models to alleviate lung, renal, cardiovascular, metabolic, cutaneous, and neural diseases as well as cancer. This article is a comprehensive review summarizing our current knowledge of the AT2R, from its discovery to its position within the RAS and its overall functions. This is followed by an in-depth look at the characteristics of the AT2R, including its structure, intracellular signaling, homo- and heterodimerization, and expression. AT2R-selective ligands, from endogenous peptides to synthetic peptides and nonpeptide molecules that are used as research tools, are discussed. Finally, we summarize the known physiological roles of the AT2R and its abundant protective effects in multiple experimental disease models and expound on AT2R ligands that are undergoing development for clinical use. The present review highlights the controversial aspects and gaps in our knowledge of this receptor and illuminates future perspectives for AT2R research. SIGNIFICANCE STATEMENT: The angiotensin AT2 receptor (AT2R) is now regarded as a fully functional and important component of the renin-angiotensin system, with the potential of exerting protective actions in a variety of diseases. This review provides an in-depth view of the AT2R, which has progressed from being an enigma to becoming a therapeutic target.
Collapse
Affiliation(s)
- U Muscha Steckelings
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert E Widdop
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Edward D Sturrock
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Lizelle Lubbe
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Tahir Hussain
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Elena Kaschina
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Thomas Unger
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Anders Hallberg
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert M Carey
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Colin Sumners
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| |
Collapse
|
13
|
Lin H, Geurts F, Hassler L, Batlle D, Mirabito Colafella KM, Denton KM, Zhuo JL, Li XC, Ramkumar N, Koizumi M, Matsusaka T, Nishiyama A, Hoogduijn MJ, Hoorn EJ, Danser AHJ. Kidney Angiotensin in Cardiovascular Disease: Formation and Drug Targeting. Pharmacol Rev 2022; 74:462-505. [PMID: 35710133 PMCID: PMC9553117 DOI: 10.1124/pharmrev.120.000236] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The concept of local formation of angiotensin II in the kidney has changed over the last 10-15 years. Local synthesis of angiotensinogen in the proximal tubule has been proposed, combined with prorenin synthesis in the collecting duct. Binding of prorenin via the so-called (pro)renin receptor has been introduced, as well as megalin-mediated uptake of filtered plasma-derived renin-angiotensin system (RAS) components. Moreover, angiotensin metabolites other than angiotensin II [notably angiotensin-(1-7)] exist, and angiotensins exert their effects via three different receptors, of which angiotensin II type 2 and Mas receptors are considered renoprotective, possibly in a sex-specific manner, whereas angiotensin II type 1 (AT1) receptors are believed to be deleterious. Additionally, internalized angiotensin II may stimulate intracellular receptors. Angiotensin-converting enzyme 2 (ACE2) not only generates angiotensin-(1-7) but also acts as coronavirus receptor. Multiple, if not all, cardiovascular diseases involve the kidney RAS, with renal AT1 receptors often being claimed to exert a crucial role. Urinary RAS component levels, depending on filtration, reabsorption, and local release, are believed to reflect renal RAS activity. Finally, both existing drugs (RAS inhibitors, cyclooxygenase inhibitors) and novel drugs (angiotensin receptor/neprilysin inhibitors, sodium-glucose cotransporter-2 inhibitors, soluble ACE2) affect renal angiotensin formation, thereby displaying cardiovascular efficacy. Particular in the case of the latter three, an important question is to what degree they induce renoprotection (e.g., in a renal RAS-dependent manner). This review provides a unifying view, explaining not only how kidney angiotensin formation occurs and how it is affected by drugs but also why drugs are renoprotective when altering the renal RAS. SIGNIFICANCE STATEMENT: Angiotensin formation in the kidney is widely accepted but little understood, and multiple, often contrasting concepts have been put forward over the last two decades. This paper offers a unifying view, simultaneously explaining how existing and novel drugs exert renoprotection by interfering with kidney angiotensin formation.
Collapse
Affiliation(s)
- Hui Lin
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Frank Geurts
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Luise Hassler
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Daniel Batlle
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Katrina M Mirabito Colafella
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Kate M Denton
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Jia L Zhuo
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Xiao C Li
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Nirupama Ramkumar
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Masahiro Koizumi
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Taiji Matsusaka
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Akira Nishiyama
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Martin J Hoogduijn
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Ewout J Hoorn
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| |
Collapse
|
14
|
Nwia SM, Li XC, Leite APDO, Hassan R, Zhuo JL. The Na +/H + Exchanger 3 in the Intestines and the Proximal Tubule of the Kidney: Localization, Physiological Function, and Key Roles in Angiotensin II-Induced Hypertension. Front Physiol 2022; 13:861659. [PMID: 35514347 PMCID: PMC9062697 DOI: 10.3389/fphys.2022.861659] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/25/2022] [Indexed: 01/29/2023] Open
Abstract
The sodium (Na+)/hydrogen (H+) exchanger 3 (NHE3) is one of the most important Na+/H+ antiporters in the small intestines of the gastrointestinal tract and the proximal tubules of the kidney. The roles of NHE3 in the regulation of intracellular pH and acid-base balance have been well established in cellular physiology using in vitro techniques. Localized primarily on the apical membranes in small intestines and proximal tubules, the key action of NHE3 is to facilitate the entry of luminal Na+ and the extrusion of intracellular H+ from intestinal and proximal tubule tubular epithelial cells. NHE3 is, directly and indirectly, responsible for absorbing the majority of ingested Na+ from small and large intestines and reabsorbing >50% of filtered Na+ in the proximal tubules of the kidney. However, the roles of NHE3 in the regulation of proximal tubular Na+ transport in the integrative physiological settings and its contributions to the basal blood pressure regulation and angiotensin II (Ang II)-induced hypertension have not been well studied previously due to the lack of suitable animal models. Recently, novel genetically modified mouse models with whole-body, kidney-specific, or proximal tubule-specific deletion of NHE3 have been generated by us and others to determine the critical roles and underlying mechanisms of NHE3 in maintaining basal body salt and fluid balance, blood pressure homeostasis, and the development of Ang II-induced hypertension at the whole-body, kidney, or proximal tubule levels. The objective of this invited article is to review, update, and discuss recent findings on the critical roles of intestinal and proximal tubule NHE3 in maintaining basal blood pressure homeostasis and their potential therapeutic implications in the development of angiotensin II (Ang II)-dependent hypertension.
Collapse
Affiliation(s)
- Sarah M. Nwia
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Xiao Chun Li
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Ana Paula de Oliveira Leite
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Rumana Hassan
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jia Long Zhuo
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States,*Correspondence: Jia Long Zhuo,
| |
Collapse
|
15
|
Kim YG, Moon JY, Oh B, Chin HJ, Kim DK, Park JH, Shin SJ, Choi BS, Lim CS, Lee SH. Pressure-Natriuresis Response Is Diminished in Old Age. Front Cardiovasc Med 2022; 9:840840. [PMID: 35252404 PMCID: PMC8889037 DOI: 10.3389/fcvm.2022.840840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Age-related alterations in renal sodium handling affect blood pressure (BP). We aimed to clarify whether the pressure-natriuresis response changes with age, leading to BP elevation. METHODS A total of 4,859 participants with normal renal function from the Korean Genome and Epidemiology Study (KoGES) and 235 patients with non-diabetic chronic kidney disease (CKD) from the ESPECIAL trial were included and divided into the younger and older groups. In ESPECIAL, participants took olmesartan from weeks 0 to 16 and were educated about a low-salt diet (LSD) from weeks 8 to 16. RESULTS In both studies, older participants showed lower estimated glomerular filtration rate (eGFR) and urine concentration index and higher albuminuria. In KoGES, BP was higher and urine sodium was lower in older participants. In ESPECIAL, diastolic BP at 0 week was lower in older participants. Olmesartan reduced BP in both groups, whereas LSD decreased systolic BP only in older participants. Urine sodium increased in younger participants but decreased in older participants after olmesartan use. In KoGES, urine sodium was correlated with BP in both groups after adjusting for age, sex, and eGFR; however, the correlation coefficient was lower in older participants. In ESPECIAL, only younger participants showed a significant positive association between systolic BP and urine sodium in multiple regression analysis. CONCLUSIONS The pressure-natriuresis response was diminished in older participants with or without CKD.
Collapse
Affiliation(s)
- Yang Gyun Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Ju-Young Moon
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Bermseok Oh
- Department of Biomedical Engineering, School of Medicine, Kyung Hee University, Seoul, South Korea
| | - Ho Jun Chin
- Division of Nephrology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Dong Ki Kim
- Division of Nephrology, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jung Hwan Park
- Division of Nephrology, Department of Internal Medicine, Konkuk University School of Medicine, Seoul, South Korea
| | - Sung Joon Shin
- Division of Nephrology, Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang, South Korea
| | - Bum Soon Choi
- Division of Nephrology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Chun Soo Lim
- Division of Nephrology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Sang Ho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University College of Medicine, Seoul, South Korea
| |
Collapse
|
16
|
Samuel CS, Bennett RG. Relaxin as an anti-fibrotic treatment: Perspectives, challenges and future directions. Biochem Pharmacol 2021; 197:114884. [PMID: 34968489 DOI: 10.1016/j.bcp.2021.114884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 02/07/2023]
Abstract
Fibrosis refers to the scarring and hardening of tissues, which results from a failed immune system-coordinated wound healing response to chronic organ injury and which manifests from the aberrant accumulation of various extracellular matrix components (ECM), primarily collagen. Despite being a hallmark of prolonged tissue damage and related dysfunction, and commonly associated with high morbidity and mortality, there are currently no effective cures for its regression. An emerging therapy that meets several criteria of an effective anti-fibrotic treatment, is the recombinant drug-based form of the human hormone, relaxin (also referred to as serelaxin, which is bioactive in several other species). This review outlines the broad anti-fibrotic and related organ-protective roles of relaxin, mainly from studies conducted in preclinical models of ageing and fibrotic disease, including its ability to ameliorate several aspects of fibrosis progression and maturation, from immune cell infiltration, pro-inflammatory and pro-fibrotic cytokine secretion, oxidative stress, organ hypertrophy, cell apoptosis, myofibroblast differentiation and ECM production, to its ability to facilitate established ECM degradation. Studies that have compared and/or combined these therapeutic effects of relaxin with current standard of care medication have also been discussed, along with the main challenges that have hindered the translation of the anti-fibrotic efficacy of relaxin to the clinic. The review then outlines the future directions as to where scientists and several pharmaceutical companies that have recognized the therapeutic potential of relaxin are working towards, to progress its development as a treatment for human patients suffering from various fibrotic diseases.
Collapse
Affiliation(s)
- Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia.
| | - Robert G Bennett
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; Department of Internal Medicine, Division of Diabetes, Endocrinology & Metabolism, University of Nebraska Medical Center, Omaha, NE 68198-4130, USA.
| |
Collapse
|
17
|
Torres-Pinzon DL, Ralph DL, Veiras LC, McDonough AA. Sex-specific adaptations to high-salt diet preserve electrolyte homeostasis with distinct sodium transporter profiles. Am J Physiol Cell Physiol 2021; 321:C897-C909. [PMID: 34613843 PMCID: PMC8616593 DOI: 10.1152/ajpcell.00282.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 02/04/2023]
Abstract
Kidneys continuously filter an enormous amount of sodium and adapt kidney Na+ reabsorption to match Na+ intake to maintain circulatory volume and electrolyte homeostasis. Males (M) respond to high-salt (HS) diet by translocating proximal tubule Na+/H+ exchanger isoform 3 (NHE3) to the base of the microvilli, reducing activated forms of the distal NaCl cotransporter (NCC) and epithelial Na+ channel (ENaC). Males (M) and females (F) on normal-salt (NS) diet present sex-specific profiles of "transporters" (cotransporters, channels, pumps, and claudins) along the nephron, e.g., F exhibit 40% lower NHE3 and 200% higher NCC abundance than M. We tested the hypothesis that adaptations to HS diet along the nephron will, likewise, exhibit sexual dimorphisms. C57BL/6J mice were fed for 15 days with 4% NaCl diet (HS) versus 0.26% NaCl diet (NS). On HS, M and F exhibited normal plasma [Na+] and [K+], similar urine volume, Na+, K+, and osmolal excretion rates normalized to body weight. In F, like M, HS lowered abundance of distal NCC, phosphorylated NCC, and cleaved (activated) forms of ENaC. The adaptations associated with achieving electrolyte homeostasis exhibit sex-dependent and independent mechanisms. Sex differences in baseline "transporters" abundance persist during HS diet, yet the fold changes during HS diet (normalized to NS) are similar along the distal nephron and collecting duct. Sex-dependent differences observed along the proximal tubule during HS show that female kidneys adapt differently from patterns reported in males, yet achieve and maintain fluid and electrolyte homeostasis.
Collapse
Affiliation(s)
- Diana L Torres-Pinzon
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Donna L Ralph
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Luciana C Veiras
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Alicia A McDonough
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| |
Collapse
|
18
|
Barsha G, Walton SL, Kwok E, Mirabito Colafella KM, Pinar AA, Hilliard Krause LM, Gaspari TA, Widdop RE, Samuel CS, Denton KM. Relaxin Attenuates Organ Fibrosis via an Angiotensin Type 2 Receptor Mechanism in Aged Hypertensive Female Rats. KIDNEY360 2021; 2:1781-1792. [PMID: 35373008 PMCID: PMC8785838 DOI: 10.34067/kid.0002722021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 09/07/2021] [Indexed: 02/04/2023]
Abstract
Background The antifibrotic effects of recombinant human relaxin (RLX) in the kidney are dependent on an interaction between its cognate receptor (RXFP1) and the angiotensin type 2 receptor (AT2R) in male models of disease. Whether RLX has therapeutic effects, which are also mediated via AT2R, in hypertensive adult and aged/reproductively senescent females is unknown. Thus, we determined whether treatment with RLX provides cardiorenal protection via an AT2R-dependent mechanism in adult and aged female stroke-prone spontaneously hypertensive rats (SHRSPs). Methods In 6-month-old (6MO) and 15-month-old ([15MO]; reproductively senescent) female SHRSP, systolic BP (SBP), GFR, and proteinuria were measured before and after 4 weeks of treatment with vehicle (Veh), RLX (0.5 mg/kg per day s.c.), or RLX+PD123319 (AT2R antagonist; 3 mg/kg per day s.c.). Aortic endothelium-dependent relaxation and fibrosis of the kidney, heart, and aorta were assessed. Results In 6MO SHRSP, RLX significantly enhanced GFR by approximately 25% (P=0.001) and reduced cardiac fibrosis (P=0.01) as compared with vehicle-treated counterparts. These effects were abolished or blunted by PD123319 coadministration. In 15MO females, RLX reduced interstitial renal (P=0.02) and aortic (P=0.003) fibrosis and lowered SBP (13±3 mm Hg; P=0.04) relative to controls. These effects were also blocked by PD123319 cotreatment (all P=0.05 versus RLX treatment alone). RLX also markedly improved vascular function by approximately 40% (P<0.001) in 15MO SHRSP, but this was not modulated by PD123319 cotreatment. Conclusions The antifibrotic and organ-protective effects of RLX, when administered to a severe model of hypertension, conferred cardiorenal protection in adult and reproductively senescent female rats to a great extent via an AT2R-mediated mechanism.
Collapse
Affiliation(s)
- Giannie Barsha
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia,Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Sarah L. Walton
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia,Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Edmund Kwok
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia,Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Katrina M. Mirabito Colafella
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia,Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Anita A. Pinar
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia,Department of Pharmacology, Monash University, Melbourne, Victoria, Australia
| | - Lucinda M. Hilliard Krause
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia,Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Tracey A. Gaspari
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia,Department of Pharmacology, Monash University, Melbourne, Victoria, Australia
| | - Robert E. Widdop
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia,Department of Pharmacology, Monash University, Melbourne, Victoria, Australia
| | - Chrishan S. Samuel
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia,Department of Pharmacology, Monash University, Melbourne, Victoria, Australia
| | - Kate M. Denton
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia,Department of Physiology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
19
|
Liao MC, Pang YC, Chang SY, Zhao XP, Chenier I, Ingelfinger JR, Chan JSD, Zhang SL. AT 2R deficiency in mice accelerates podocyte dysfunction in diabetic progeny in a sex-dependent manner. Diabetologia 2021; 64:2108-2121. [PMID: 34047808 DOI: 10.1007/s00125-021-05483-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/11/2021] [Indexed: 10/21/2022]
Abstract
AIMS/HYPOTHESIS The angiotensin II receptor type 2 (AT2R) may be a potential therapeutic target for the treatment of hypertension and chronic kidney disease (CKD). The expression and function of AT2R in the vasculature and kidney appear sexually dimorphic. We hypothesised that Agtr2 knockout dams (AT2RKO) with gestational diabetes would program their offspring for subsequent hypertension and CKD in a sex-dependent manner. METHODS Age- and sex-matched offspring of non-diabetic and diabetic dams of wild-type (WT) and AT2RKO mice were followed from 4 to 20 weeks of age and were monitored for development of hypertension and nephropathy; a mouse podocyte cell line (mPOD) was also studied. RESULTS Body weight was progressively lower in female compared with male offspring throughout the lifespan. Female but not male offspring from diabetic AT2RKO dams developed insulin resistance. Compared with the offspring of non-diabetic dams, the progeny of diabetic dams had developed more hypertension and nephropathy (apparent glomerulosclerosis with podocyte loss) at 20 weeks of age; this programming was more pronounced in the offspring of AT2RKO diabetic dams, particularly female AT2RKO progeny. Female AT2RKO offspring had lower basal ACE2 glomerular expression, resulting in podocyte loss. The aberrant ACE2/ACE ratio was far more diminished in glomeruli of female progeny of diabetic AT2RKO dams than in male progeny. Knock-down of Agtr2 in mPODs confirmed the in vivo data. CONCLUSIONS/INTERPRETATION AT2R deficiency accelerated kidney programming in female progeny of diabetic dams, possibly due to loss of protective effects of ACE2 expression in the kidney.
Collapse
Affiliation(s)
- Min-Chun Liao
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Yu-Chao Pang
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Shiao-Ying Chang
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Xin-Ping Zhao
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Isabelle Chenier
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Julie R Ingelfinger
- Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - John S D Chan
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Shao-Ling Zhang
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.
| |
Collapse
|
20
|
Bhargava A, Arnold AP, Bangasser DA, Denton KM, Gupta A, Hilliard Krause LM, Mayer EA, McCarthy M, Miller WL, Raznahan A, Verma R. Considering Sex as a Biological Variable in Basic and Clinical Studies: An Endocrine Society Scientific Statement. Endocr Rev 2021; 42:219-258. [PMID: 33704446 PMCID: PMC8348944 DOI: 10.1210/endrev/bnaa034] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Indexed: 02/08/2023]
Abstract
In May 2014, the National Institutes of Health (NIH) stated its intent to "require applicants to consider sex as a biological variable (SABV) in the design and analysis of NIH-funded research involving animals and cells." Since then, proposed research plans that include animals routinely state that both sexes/genders will be used; however, in many instances, researchers and reviewers are at a loss about the issue of sex differences. Moreover, the terms sex and gender are used interchangeably by many researchers, further complicating the issue. In addition, the sex or gender of the researcher might influence study outcomes, especially those concerning behavioral studies, in both animals and humans. The act of observation may change the outcome (the "observer effect") and any experimental manipulation, no matter how well-controlled, is subject to it. This is nowhere more applicable than in physiology and behavior. The sex of established cultured cell lines is another issue, in addition to aneuploidy; chromosomal numbers can change as cells are passaged. Additionally, culture medium contains steroids, growth hormone, and insulin that might influence expression of various genes. These issues often are not taken into account, determined, or even considered. Issues pertaining to the "sex" of cultured cells are beyond the scope of this Statement. However, we will discuss the factors that influence sex and gender in both basic research (that using animal models) and clinical research (that involving human subjects), as well as in some areas of science where sex differences are routinely studied. Sex differences in baseline physiology and associated mechanisms form the foundation for understanding sex differences in diseases pathology, treatments, and outcomes. The purpose of this Statement is to highlight lessons learned, caveats, and what to consider when evaluating data pertaining to sex differences, using 3 areas of research as examples; it is not intended to serve as a guideline for research design.
Collapse
Affiliation(s)
- Aditi Bhargava
- Center for Reproductive Sciences, San Francisco, CA, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, CA, USA
| | - Arthur P Arnold
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Arpana Gupta
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lucinda M Hilliard Krause
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, CA, USA
| | - Margaret McCarthy
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Walter L Miller
- Center for Reproductive Sciences, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institutes of Mental Health, Intramural Research Program, Bethesda, MD, USA
| | - Ragini Verma
- Diffusion and Connectomics In Precision Healthcare Research (DiCIPHR) lab, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
21
|
Barsha G, Mirabito Colafella KM, Walton SL, Gaspari TA, Spizzo I, Pinar AA, Hilliard Krause LM, Widdop RE, Samuel CS, Denton KM. In Aged Females, the Enhanced Pressor Response to Angiotensin II Is Attenuated By Estrogen Replacement via an Angiotensin Type 2 Receptor-Mediated Mechanism. Hypertension 2021; 78:128-137. [PMID: 33966450 DOI: 10.1161/hypertensionaha.121.17164] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Giannie Barsha
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Physiology (G.B., KM.M.C., S.L.W., L.M.H.K., K.M.D.), Monash University, Melbourne, Victoria, Australia
| | - Katrina M Mirabito Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Physiology (G.B., KM.M.C., S.L.W., L.M.H.K., K.M.D.), Monash University, Melbourne, Victoria, Australia
| | - Sarah L Walton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Physiology (G.B., KM.M.C., S.L.W., L.M.H.K., K.M.D.), Monash University, Melbourne, Victoria, Australia
| | - Tracey A Gaspari
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Pharmacology (T.A.G., I.S., A.A.P., R.E.W., C.S.S.), Monash University, Melbourne, Victoria, Australia
| | - Iresha Spizzo
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Pharmacology (T.A.G., I.S., A.A.P., R.E.W., C.S.S.), Monash University, Melbourne, Victoria, Australia
| | - Anita A Pinar
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Pharmacology (T.A.G., I.S., A.A.P., R.E.W., C.S.S.), Monash University, Melbourne, Victoria, Australia
| | - Lucinda M Hilliard Krause
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Physiology (G.B., KM.M.C., S.L.W., L.M.H.K., K.M.D.), Monash University, Melbourne, Victoria, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Pharmacology (T.A.G., I.S., A.A.P., R.E.W., C.S.S.), Monash University, Melbourne, Victoria, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Pharmacology (T.A.G., I.S., A.A.P., R.E.W., C.S.S.), Monash University, Melbourne, Victoria, Australia
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Physiology (G.B., KM.M.C., S.L.W., L.M.H.K., K.M.D.), Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
22
|
Fatima N, Patel SN, Hussain T. Angiotensin II Type 2 Receptor: A Target for Protection Against Hypertension, Metabolic Dysfunction, and Organ Remodeling. Hypertension 2021; 77:1845-1856. [PMID: 33840201 PMCID: PMC8115429 DOI: 10.1161/hypertensionaha.120.11941] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The renin-angiotensin system is of vital significance not only in the maintenance of blood pressure but also because of its role in the pathophysiology of different organ systems in the body. Of the 2 Ang II (angiotensin II) receptors, the AT1R (Ang II type 1 receptor) has been extensively studied for its role in mediating the classical functions of Ang II, including vasoconstriction, stimulation of renal tubular sodium reabsorption, hormonal secretion, cell proliferation, inflammation, and oxidative stress. The other receptor, AT2R (Ang II type 2 receptor), is abundantly expressed in both immune and nonimmune cells in fetal tissue. However, its expression is increased under pathological conditions in adult tissues. The role of AT2R in counteracting AT1R function has been discussed in the past 2 decades. However, with the discovery of the nonpeptide agonist C21, the significance of AT2R in various pathologies such as obesity, hypertension, and kidney diseases have been examined. This review focuses on the most recent findings on the beneficial effects of AT2R by summarizing both gene knockout studies as well as pharmacological studies, specifically highlighting its importance in blood pressure regulation, obesity/metabolism, organ protection, and relevance in the treatment of coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Naureen Fatima
- From the Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, TX
| | - Sanket N Patel
- From the Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, TX
| | - Tahir Hussain
- From the Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, TX
| |
Collapse
|
23
|
Bovée DM, Cuevas CA, Zietse R, Danser AHJ, Mirabito Colafella KM, Hoorn EJ. Salt-sensitive hypertension in chronic kidney disease: distal tubular mechanisms. Am J Physiol Renal Physiol 2020; 319:F729-F745. [DOI: 10.1152/ajprenal.00407.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD) causes salt-sensitive hypertension that is often resistant to treatment and contributes to the progression of kidney injury and cardiovascular disease. A better understanding of the mechanisms contributing to salt-sensitive hypertension in CKD is essential to improve these outcomes. This review critically explores these mechanisms by focusing on how CKD affects distal nephron Na+ reabsorption. CKD causes glomerulotubular imbalance with reduced proximal Na+ reabsorption and increased distal Na+ delivery and reabsorption. Aldosterone secretion further contributes to distal Na+ reabsorption in CKD and is not only mediated by renin and K+ but also by metabolic acidosis, endothelin-1, and vasopressin. CKD also activates the intrarenal renin-angiotensin system, generating intratubular angiotensin II to promote distal Na+ reabsorption. High dietary Na+ intake in CKD contributes to Na+ retention by aldosterone-independent activation of the mineralocorticoid receptor mediated through Rac1. High dietary Na+ also produces an inflammatory response mediated by T helper 17 cells and cytokines increasing distal Na+ transport. CKD is often accompanied by proteinuria, which contains plasmin capable of activating the epithelial Na+ channel. Thus, CKD causes both local and systemic changes that together promote distal nephron Na+ reabsorption and salt-sensitive hypertension. Future studies should address remaining knowledge gaps, including the relative contribution of each mechanism, the influence of sex, differences between stages and etiologies of CKD, and the clinical relevance of experimentally identified mechanisms. Several pathways offer opportunities for intervention, including with dietary Na+ reduction, distal diuretics, renin-angiotensin system inhibitors, mineralocorticoid receptor antagonists, and K+ or H+ binders.
Collapse
Affiliation(s)
- Dominique M. Bovée
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
- Division of Vascular Medicine, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Catharina A. Cuevas
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robert Zietse
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A. H. Jan Danser
- Division of Vascular Medicine, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Katrina M. Mirabito Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Ewout J. Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
24
|
Kemp BA, Howell NL, Keller SR, Gildea JJ, Shao W, Navar LG, Carey RM. Defective Renal Angiotensin III and AT 2 Receptor Signaling in Prehypertensive Spontaneously Hypertensive Rats. J Am Heart Assoc 2020; 8:e012016. [PMID: 31039659 PMCID: PMC6512109 DOI: 10.1161/jaha.119.012016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Previous studies demonstrated that angiotensin (Ang) III, not Ang II, is the predominant endogenous agonist for Ang type‐2 receptor (AT2R)‐induced natriuresis in normal rats, and that hypertensive 12‐week‐old spontaneously hypertensive rats (SHR) lack natriuretic responses to Ang III. This study tested whether prehypertensive SHR already have defective Ang III‐induced natriuresis and determined possible mechanisms. Methods and Results Female and male normotensive 4‐week‐old SHR and Wistar Kyoto rats were studied after 24‐hour systemic AT1R blockade. Left kidneys received 30 minute renal interstitial infusions of vehicle followed by Ang III (3.5, 7.0, 14, and 28 nmol/kg per min; each dose for 30 minutes). Right kidneys received vehicle infusions. In 4‐week‐old Wistar Kyoto rats, renal interstitial Ang III increased urine sodium (Na+) excretion but failed to induce natriuresis in 4‐week‐old SHR. Renal Ang III levels were similar between Wistar Kyoto rats and SHR, making increased Ang III degradation as a possible cause for defective natriuresis in SHR unlikely. In Wistar Kyoto rats, renal interstitial Ang III induced translocation of AT2Rs to apical plasma membranes of renal proximal tubule cells. Simultaneously, Ang III induced retraction of the major Na+ transporter Na+‐H+ exchanger‐3 (NHE‐3) from apical membranes and internalization of Na+/K+ATPase (NKA) from basolateral membranes of renal proximal tubule cells. Consistent with NHE‐3 and NKA retraction, Ang III increased pSer552‐NHE‐3 and decreased pSer23‐NKA. In contrast, in SHR, intrarenal Ang III failed to induce AT2R translocation, NHE‐3 or NKA retraction, pSer552‐NHE‐3 phosphorylation, or pSer23‐NKA dephosphorylation. Conclusions These results indicate impaired Ang III/AT2R signaling as a possible primary defect in prehypertensive SHR. See Editorial Zhuo and Li
Collapse
Affiliation(s)
- Brandon A Kemp
- 1 Division of Endocrinology and Metabolism Department of Medicine University of Virginia School of Medicine Charlottesville VA
| | - Nancy L Howell
- 1 Division of Endocrinology and Metabolism Department of Medicine University of Virginia School of Medicine Charlottesville VA
| | - Susanna R Keller
- 1 Division of Endocrinology and Metabolism Department of Medicine University of Virginia School of Medicine Charlottesville VA
| | - John J Gildea
- 2 Department of Pathology University of Virginia School of Medicine Charlottesville VA
| | - Weijian Shao
- 3 Department of Physiology and Hypertension and Renal Center Tulane University School of Medicine New Orleans LA
| | - Luis Gabriel Navar
- 3 Department of Physiology and Hypertension and Renal Center Tulane University School of Medicine New Orleans LA
| | - Robert M Carey
- 1 Division of Endocrinology and Metabolism Department of Medicine University of Virginia School of Medicine Charlottesville VA
| |
Collapse
|
25
|
Hu R, McDonough AA, Layton AT. Sex differences in solute transport along the nephrons: effects of Na + transport inhibition. Am J Physiol Renal Physiol 2020; 319:F487-F505. [PMID: 32744084 DOI: 10.1152/ajprenal.00240.2020] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Each day, ~1.7 kg of NaCl and 180 liters of water are reabsorbed by nephron segments in humans, with urinary excretion fine tuned to meet homeostatic requirements. These tasks are coordinated by a spectrum of renal Na+ transporters and channels. The goal of the present study was to investigate the extent to which inhibitors of transepithelial Na+ transport (TNa) along the nephron alter urinary solute excretion and how those effects may vary between male and female subjects. To accomplish that goal, we developed sex-specific multinephron models that represent detailed transcellular and paracellular transport processes along the nephrons of male and female rat kidneys. We simulated inhibition of Na+/H+ exchanger 3 (NHE3), bumetanide-sensitive Na+-K+-2Cl- cotransporter (NKCC2), Na+-Cl- cotransporter (NCC), and amiloride-sensitive epithelial Na+ channel (ENaC). NHE3 inhibition simulations predicted a substantially reduced proximal tubule TNa, and NKCC2 inhibition substantially reduced thick ascending limb TNa. Both gave rise to diuresis, natriuresis, and kaliuresis, with those effects stronger in female rats. While NCC inhibition was predicted to have only minor impact on renal TNa, it nonetheless had a notable effect of enhancing excretion of Na+, K+, and Cl-, particularly in female rats. Inhibition of ENaC was predicted to have opposite effects on the excretion of Na+ (increased) and K+ (decreased) and to have only a minor impact on whole kidney TNa. Unlike inhibition of other transporters, ENaC inhibition induced stronger natriuresis and diuresis in male rats than female rats. Overall, model predictions agreed well with measured changes in Na+ and K+ excretion in response to diuretics and Na+ transporter mutations.
Collapse
Affiliation(s)
- Rui Hu
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Alicia A McDonough
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Anita T Layton
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada.,Department of Biology, Cheriton School of Computer Science, and School of Pharmacology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
26
|
Veiras LC, McFarlin BE, Ralph DL, Buncha V, Prescott J, Shirvani BS, McDonough JC, Ha D, Giani J, Gurley SB, Mamenko M, McDonough AA. Electrolyte and transporter responses to angiotensin II induced hypertension in female and male rats and mice. Acta Physiol (Oxf) 2020; 229:e13448. [PMID: 31994810 DOI: 10.1111/apha.13448] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/15/2022]
Abstract
AIM Sexual dimorphisms are evident along the nephron: Females (F) exhibit higher ratios of renal distal to proximal Na+ transporters' abundance, greater lithium clearance (CLi ) more rapid natriuresis in response to saline infusion and lower plasma [K+ ] vs. males (M). During angiotensin II infusion hypertension (AngII-HTN) M exhibit distal Na+ transporter activation, lower proximal and medullary loop transporters, blunted natriuresis in response to saline load, and reduced plasma [K+ ]. This study aimed to determine whether responses of F to AngII-HTN mimicked those in M or were impacted by sexual dimorphisms evident at baseline. METHODS Sprague Dawley rats and C57BL/6 mice were AngII infused via osmotic minipumps 2 and 3 weeks, respectively, and assessed by metabolic cage collections, tail-cuff sphygmomanometer, semi-quantitative immunoblotting of kidney and patch-clamp electrophysiology. RESULTS In F rats, AngII-infusion increased BP to 190 mm Hg, increased phosphorylation of cortical NKCC2, NCC and cleavage of ENaC two to threefold, increased ENaC channel activity threefold and aldosterone 10-fold. K+ excretion increased and plasma [K+ ] decreased. Evidence of natriuresis in F included increased urine Na+ excretion and CLi , and decreased medullary NHE3, NKCC2 and Na,K-ATPase abundance. In C57BL/6 mice, AngII-HTN increased abundance of distal Na+ transporters, suppressed proximal-medullary transporters and reduced plasma [K+ ] in both F and M. CONCLUSION Despite baseline sexual dimorphisms, AngII-HTN provokes similar increases in BP, aldosterone, distal transporters, ENaC channel activation and K+ loss accompanied by similar suppression of proximal and loop Na+ transporters, natriuresis and diuresis in females and males.
Collapse
Affiliation(s)
- Luciana C. Veiras
- Department of Physiology and Neuroscience Keck School of Medicine of USC Los Angeles CA USA
- Department of Biomedical Sciences Cedars‐Sinai Medical Center Los Angeles CA USA
| | - Brandon E. McFarlin
- Department of Physiology and Neuroscience Keck School of Medicine of USC Los Angeles CA USA
| | - Donna L. Ralph
- Department of Physiology and Neuroscience Keck School of Medicine of USC Los Angeles CA USA
| | - Vadym Buncha
- Department of Physiology Medical College of Georgia at Augusta University Augusta GA USA
| | - Jessica Prescott
- Department of Physiology and Neuroscience Keck School of Medicine of USC Los Angeles CA USA
| | - Borna S. Shirvani
- Department of Physiology and Neuroscience Keck School of Medicine of USC Los Angeles CA USA
| | - Jillian C. McDonough
- Department of Physiology and Neuroscience Keck School of Medicine of USC Los Angeles CA USA
| | - Darren Ha
- Department of Physiology and Neuroscience Keck School of Medicine of USC Los Angeles CA USA
| | - Jorge Giani
- Department of Biomedical Sciences Cedars‐Sinai Medical Center Los Angeles CA USA
| | - Susan B. Gurley
- Division of Nephrology and Hypertension Oregon Health and Science University Portland OR USA
| | - Mykola Mamenko
- Department of Physiology Medical College of Georgia at Augusta University Augusta GA USA
| | - Alicia A. McDonough
- Department of Physiology and Neuroscience Keck School of Medicine of USC Los Angeles CA USA
| |
Collapse
|
27
|
Wang C, Pinar AA, Widdop RE, Hossain MA, Bathgate RAD, Denton KM, Kemp-Harper BK, Samuel CS. The anti-fibrotic actions of relaxin are mediated through AT 2 R-associated protein phosphatases via RXFP1-AT 2 R functional crosstalk in human cardiac myofibroblasts. FASEB J 2020; 34:8217-8233. [PMID: 32297670 DOI: 10.1096/fj.201902506r] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 12/19/2022]
Abstract
Fibrosis is a hallmark of several cardiovascular diseases. The relaxin family peptide receptor 1 (RXFP1) agonist, relaxin, has rapidly occurring anti-fibrotic actions which are mediated through RXFP1 and angiotensin II receptor crosstalk on renal and cardiac myofibroblasts. Here, we investigated whether this would allow relaxin to indirectly activate angiotensin II type 2 receptor (AT2 R)-specific signal transduction in primary human cardiac myofibroblasts (HCMFs). The anti-fibrotic effects of recombinant human relaxin (RLX; 16.8 nM) or the AT2 R-agonist, Compound 21 (C21; 1 μM), were evaluated in TGF-β1-stimulated HCMFs, in the absence or presence of an RXFP1 antagonist (1 μM) or AT2 R antagonist (0.1 μM) to confirm RXFP1-AT2 R crosstalk. Competition binding for RXFP1 was determined. Western blotting was performed to determine which AT2 R-specific protein phosphatases were expressed by HCMFs; then, the anti-fibrotic effects of RLX and/or C21 were evaluated in the absence or presence of pharmacological inhibition (NSC95397 (1 μM) for MKP-1; okadaic acid (10 nM) for PP2A) or siRNA-knockdown of these phosphatases after 72 hours. The RLX- or C21-induced increase in ERK1/2 and nNOS phosphorylation, and decrease in α-SMA (myofibroblast differentiation) and collagen-I expression by HCMFs was abrogated by pharmacological blockade of RXFP1 or the AT2 R, confirming RXFP1-AT2 R crosstalk in these cells. HCMFs were found to express AT2 R-dependent MKP-1 and PP2A phosphatases, while pharmacological blockade or siRNA-knockdown of either phosphatase also abolished RLX and/or C21 signal transduction in HCMFs (all P < .05 vs RLX or C21 alone). These findings demonstrated that RLX can indirectly activate AT2 R-dependent phosphatase activity in HCMFs by signaling through RXFP1-AT2 R crosstalk, which have important therapeutic implications for its anti-fibrotic actions.
Collapse
Affiliation(s)
- Chao Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Anita A Pinar
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Mohammed A Hossain
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Ross A D Bathgate
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Barbara K Kemp-Harper
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Pharmacology, Monash University, Clayton, VIC, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
28
|
Assersen KB, Sumners C, Steckelings UM. The Renin-Angiotensin System in Hypertension, a Constantly Renewing Classic: Focus on the Angiotensin AT 2-Receptor. Can J Cardiol 2020; 36:683-693. [PMID: 32389341 DOI: 10.1016/j.cjca.2020.02.095] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 02/07/2023] Open
Abstract
It is common knowledge that the renin-angiotensin system (RAS), in particular angiotensin II acting through the angiotensin AT1-receptor (AT1R), is pivotal for the regulation of blood pressure (BP) and extracellular volume. More recent findings have revealed that the RAS is far more complex than initially thought and that it harbours additional mediators and receptors, which are able to counteract and thereby fine-tune AT1R-mediated actions. This review will focus on the angiotensin AT2-receptor (AT2R), which is one of the "counter-regulatory" receptors within the RAS. It will review and discuss data related to the role of the AT2R in regulation of BP and focus on the following 3 questions: Do peripheral AT2R have an impact on BP regulation, and, if so, does this effect become apparent only under certain conditions? Are central nervous system AT2R involved in regulation of BP, and, if so, which brain areas are involved and what are the mechanisms? Does dysfunction of AT2R contribute to the pathogenesis of hypertension in preeclampsia?
Collapse
Affiliation(s)
- Kasper B Assersen
- Institute for Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Colin Sumners
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - U Muscha Steckelings
- Institute for Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
29
|
Gonçalves GD, Walton SL, Gazzard SE, van der Wolde J, Mathias PCF, Moritz KM, Cullen-McEwen LA, Bertram JF. Maternal hypoxia developmentally programs low podocyte endowment in male, but not female offspring. Anat Rec (Hoboken) 2020; 303:2668-2678. [PMID: 31984678 DOI: 10.1002/ar.24369] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/02/2019] [Accepted: 12/08/2019] [Indexed: 11/07/2022]
Abstract
Fetal hypoxia is a common complication of pregnancy. We have previously reported that maternal hypoxia in late gestation in mice gives rise to male offspring with reduced nephron number, while females have normal nephron number. Male offspring later develop proteinuria and renal pathology, including glomerular pathology, whereas female offspring are unaffected. Given the central role of podocyte depletion in glomerular and renal pathology, we examined whether maternal hypoxia resulted in low podocyte endowment in offspring. Pregnant CD1 mice were allocated at embryonic day 14.5 to normoxic (21% oxygen) or hypoxic (12% oxygen) conditions. At postnatal day 21, kidneys from mice were immersion fixed, and one mid-hilar slice per kidney was immunostained with antibodies directed against p57 and synaptopodin for podocyte identification. Slices were cleared and imaged with a multiphoton microscope for podometric analysis. Male hypoxic offspring had significantly lower birth weight, nephron number, and podocyte endowment than normoxic male offspring (podocyte number; normoxic 62.86 ± 2.26 podocytes per glomerulus, hypoxic 53.38 ± 2.25; p < .01, mean ± SEM). In contrast, hypoxic female offspring had low birth weight but their nephron and podocyte endowment was the same as normoxic female offspring (podocyte number; normoxic 62.38 ± 1.86 podocytes per glomerulus, hypoxic 61.81 ± 1.80; p = .88). To the best of our knowledge, this is the first report of developmentally programmed low podocyte endowment. Given the well-known association between podocyte depletion in adulthood and glomerular pathology, we postulate that podocyte endowment may place offspring at risk of renal disease in adulthood, and explain the greater vulnerability of male offspring.
Collapse
Affiliation(s)
- Gessica D Gonçalves
- Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia.,Biological Science Program, Department of Biotechnology, Genetics and Cellular Biology, State University of Maringá, Maringá, Brazil
| | - Sarah L Walton
- School of Biomedical Sciences and Child Health Research Centre, The University of Queensland, Brisbane, Australia.,Cardiovascular Disease Program, and Department of Physiology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Sarah E Gazzard
- Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - James van der Wolde
- Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Paulo C F Mathias
- Biological Science Program, Department of Biotechnology, Genetics and Cellular Biology, State University of Maringá, Maringá, Brazil
| | - Karen M Moritz
- School of Biomedical Sciences and Child Health Research Centre, The University of Queensland, Brisbane, Australia
| | - Luise A Cullen-McEwen
- Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - John F Bertram
- Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| |
Collapse
|
30
|
Wang Y, Han L, Shen M, Jones ES, Spizzo I, Walton SL, Denton KM, Gaspari TA, Samuel CS, Widdop RE. Serelaxin and the AT 2 Receptor Agonist CGP42112 Evoked a Similar, Nonadditive, Cardiac Antifibrotic Effect in High Salt-Fed Mice That Were Refractory to Candesartan Cilexetil. ACS Pharmacol Transl Sci 2020; 3:76-87. [PMID: 32259090 DOI: 10.1021/acsptsci.9b00095] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Indexed: 12/29/2022]
Abstract
Fibrosis is involved in the majority of cardiovascular diseases and is a key contributor to end-organ dysfunction. In the current study, the antifibrotic effects of recombinant human relaxin-2 (serelaxin; RLX) and/or the AT2R agonist CGP42112 (CGP) were compared with those of the established AT1R antagonist, candesartan cilexetil (CAND), in a high salt-induced cardiac fibrosis model. High salt (HS; 5%) for 8 weeks did not increase systolic blood pressure in male FVB/N mice, but CAND treatment alone significantly reduced systolic blood pressure from HS-induced levels. HS significantly increased cardiac interstitial fibrosis, which was reduced by either RLX and/or CGP, which were not additive under the current experimental conditions, while CAND failed to reduce HS-induced cardiac fibrosis. The antifibrotic effects induced by RLX and/or CGP were associated with reduced myofibroblast differentiation. Additionally, all treatments inhibited the HS-induced elevation in tissue inhibitor of matrix metalloproteinases-1, together with trends for increased MMP-13 expression, that collectively would favor collagen degradation. Furthermore, these antifibrotic effects were associated with reduced cardiac inflammation. Collectively, these results highlight that either RXFP1 or AT2R stimulation represents novel therapeutic strategies to target fibrotic conditions, particularly in HS states that may be refractory to AT1R blockade.
Collapse
Affiliation(s)
- Yan Wang
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Lei Han
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Matthew Shen
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Emma S Jones
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Iresha Spizzo
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Sarah L Walton
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Kate M Denton
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Tracey A Gaspari
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| |
Collapse
|
31
|
FGF-21 ameliorates essential hypertension of SHR via baroreflex afferent function. Brain Res Bull 2019; 154:9-20. [PMID: 31626954 DOI: 10.1016/j.brainresbull.2019.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/29/2019] [Accepted: 10/11/2019] [Indexed: 12/28/2022]
Abstract
Hypertension is a common complication of metabolic abnormalities associated with cardiovascular system and characterized by sexual dimorphism in mammals. Fibroblast growth factor-21 (FGF-21) plays a critical role in metabolic-disorder related hypertension through the afferent loop of baroreflex. However, the gender difference in FGF-21-mediated blood pressure (BP) regulation via sexual dimorphic expression of FGFRs in the nodose (NG) and nucleus tractus solitarius (NTS) were not elucidated in physiological and genomic form of hypertension. The gene and protein expression of FGFRs were tested by qRT-PCR, immunoblotting and immunostaining; the serum level of FGF21 was tested using ELISA; The BP was monitored while FGF21 was nodose microinjected. The results showed that more potent BP reduction was confirmed in female vs. male rats by nodose microinjection of rhFGF-21 along with higher expression of FGFR2 and FGFR4 in the nodose compared with age-match male and ovariectomized (OVX) rats, rather than other receptor subtypes, which is consistent well with immunohistochemical analysis. Additionally, serum FGF-21 was significantly higher in female-WKY, and this level of FGF-21 was dramatically declined in spontaneous hypertensive rats (SHR) with significant down-regulation of FGFR1/R4 for male-SHR and FGFR2/FGFR4 for female-SHR, respectively. Apparently, high BP of SHR of either sex could be reduced by rhFGF-21 nodose microinjection. These data extends our current understanding that sexual-specific distribution/expression of FGF-21/FGFRs is likely to contribute at least partially to sexual dimorphism of baroreflex afferent function on BP regulation in rats. FGF-21-mdiated BP reduction sheds new light on clinical management of primary/genomic form of hypertension.
Collapse
|
32
|
Chow BSM, Kocan M, Shen M, Wang Y, Han L, Chew JY, Wang C, Bosnyak S, Mirabito-Colafella KM, Barsha G, Wigg B, Johnstone EKM, Hossain MA, Pfleger KDG, Denton KM, Widdop RE, Summers RJ, Bathgate RAD, Hewitson TD, Samuel CS. AT1R-AT2R-RXFP1 Functional Crosstalk in Myofibroblasts: Impact on the Therapeutic Targeting of Renal and Cardiac Fibrosis. J Am Soc Nephrol 2019; 30:2191-2207. [PMID: 31511361 DOI: 10.1681/asn.2019060597] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/29/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Recombinant human relaxin-2 (serelaxin), which has organ-protective actions mediated via its cognate G protein-coupled receptor relaxin family peptide receptor 1 (RXFP1), has emerged as a potential agent to treat fibrosis. Studies have shown that serelaxin requires the angiotensin II (AngII) type 2 receptor (AT2R) to ameliorate renal fibrogenesis in vitro and in vivo. Whether its antifibrotic actions are affected by modulation of the AngII type 1 receptor (AT1R), which is expressed on myofibroblasts along with RXFP1 and AT2R, is unknown. METHODS We examined the signal transduction mechanisms of serelaxin when applied to primary rat renal and human cardiac myofibroblasts in vitro, and in three models of renal- or cardiomyopathy-induced fibrosis in vivo. RESULTS The AT1R blockers irbesartan and candesartan abrogated antifibrotic signal transduction of serelaxin via RXFP1 in vitro and in vivo. Candesartan also ameliorated serelaxin's antifibrotic actions in the left ventricle of mice with cardiomyopathy, indicating that candesartan's inhibitory effects were not confined to the kidney. We also demonstrated in a transfected cell system that serelaxin did not directly bind to AT1Rs but that constitutive AT1R-RXFP1 interactions could form. To potentially explain these findings, we also demonstrated that renal and cardiac myofibroblasts expressed all three receptors and that antagonists acting at each receptor directly or allosterically blocked the antifibrotic effects of either serelaxin or an AT2R agonist (compound 21). CONCLUSIONS These findings have significant implications for the concomitant use of RXFP1 or AT2R agonists with AT1R blockers, and suggest that functional interactions between the three receptors on myofibroblasts may represent new targets for controlling fibrosis progression.
Collapse
Affiliation(s)
- Bryna S M Chow
- Florey Institute of Neuroscience and Mental Health.,Department of Biochemistry and Molecular Biology, and
| | - Martina Kocan
- Florey Institute of Neuroscience and Mental Health.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Matthew Shen
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Yan Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Lei Han
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Jacqueline Y Chew
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Chao Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Sanja Bosnyak
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia.,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Katrina M Mirabito-Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Giannie Barsha
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Belinda Wigg
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Elizabeth K M Johnstone
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | | | - Kevin D G Pfleger
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia.,Department of Pharmacology and Therapeutics, ARC Centre for Personalised Therapeutic Technologies, Melbourne, Australia; and.,Dimerix Limited, Nedlands, Western Australia, Australia
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia.,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Ross A D Bathgate
- Florey Institute of Neuroscience and Mental Health.,Department of Biochemistry and Molecular Biology, and
| | - Tim D Hewitson
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Chrishan S Samuel
- Department of Biochemistry and Molecular Biology, and .,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| |
Collapse
|
33
|
Syme C, Shin J, Richer L, Gaudet D, Paus T, Pausova Z. Sex Differences in Blood Pressure Hemodynamics in Middle-Aged Adults With Overweight and Obesity. Hypertension 2019; 74:407-412. [DOI: 10.1161/hypertensionaha.119.13058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
High blood pressure (BP) is the strongest modifiable risk factor for cardiovascular disease. Overweight/obesity is a major risk factor of high BP. Multiple sex differences exist in mechanistic pathways that increase BP in overweight/obesity. They may result in a sex-specific pattern of BP hemodynamics—males and females may vary in the relative contributions of stroke volume, total peripheral resistance (TPR), and heart rate to higher BP. We investigated this possibility in a population-based sample of middle-aged adults (36–65 years). The total sample (n=618) included 289 males and 329 females; 79% of males and 66% of females were overweight. In all, we measured BP, stroke volume, TPR, and heart rate beat-by-beat during a 52-minute protocol that included changes in posture and mental stress. We assessed the relative contributions of stroke volume, TPR, and heart rate to BP at each minute of the protocol. We observed marked sex differences in BP hemodynamics in overweight/obese individuals: the main determinant of higher BP was TPR in males (49% versus only 35% in females,
P
=0.008), whereas it was stroke volume in females (51% versus only 35% in males,
P
=0.006). These sex differences were most apparent when standing or sitting at rest. No such differences were seen in normal-weight individuals in whom the main determinant of higher BP was TPR in both sexes. Our study suggests that, in middle-aged adults, marked sex differences exist in BP hemodynamics, contributing to high BP in overweight/obese but not normal-weight individuals. As such, this study may contribute to precision medicine in hypertension.
Collapse
Affiliation(s)
- Catriona Syme
- From the Hospital for Sick Children (C.S., J.S., Z.P.), University of Toronto, Canada
- Departments of Physiology and Nutritional Sciences (C.S., J.S., Z.P.), University of Toronto, Canada
| | - Jean Shin
- From the Hospital for Sick Children (C.S., J.S., Z.P.), University of Toronto, Canada
- Departments of Physiology and Nutritional Sciences (C.S., J.S., Z.P.), University of Toronto, Canada
| | - Louis Richer
- Department of Health Sciences Université du Québec à Chicoutimi, Canada (L.R.)
| | - Daniel Gaudet
- Community Genomic Centre, Université de Montréal, Chicoutimi, Canada (D.G.)
| | - Tomáš Paus
- Departments of Psychology and Psychiatry (T.P.), University of Toronto, Canada
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Canada (T.P.)
| | - Zdenka Pausova
- From the Hospital for Sick Children (C.S., J.S., Z.P.), University of Toronto, Canada
- Departments of Physiology and Nutritional Sciences (C.S., J.S., Z.P.), University of Toronto, Canada
| |
Collapse
|
34
|
Li XC, Zhu D, Chen X, Zheng X, Zhao C, Zhang J, Soleimani M, Rubera I, Tauc M, Zhou X, Zhuo JL. Proximal Tubule-Specific Deletion of the NHE3 (Na +/H + Exchanger 3) in the Kidney Attenuates Ang II (Angiotensin II)-Induced Hypertension in Mice. Hypertension 2019; 74:526-535. [PMID: 31352824 DOI: 10.1161/hypertensionaha.119.13094] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The present study directly tested the hypothesis that the NHE3 (Na+/H+ exchanger 3) in the proximal tubules of the kidney is required for the development of Ang II (angiotensin II)-induced hypertension using PT-Nhe3-/- (proximal tubule-specific NHE3 knockout) mice. Specifically, PT-Nhe3-/- mice were generated using the SGLT2-Cre/Nhe3loxlox approach, whereas Ang II-induced hypertension was studied in 12 groups (n=5-12 per group) of adult male and female wild-type (WT) and PT-Nhe3-/- mice. Under basal conditions, systolic blood pressure, diastolic blood pressure, and mean arterial blood pressure were significantly lower in male and female PT-Nhe3-/- than WT mice (P<0.01). A high pressor, 1.5 mg/kg per day, intraperitoneal or a slow pressor dose of Ang II, 0.5 mg/kg per day, intraperitoneal for 2 weeks significantly increased systolic blood pressure, diastolic blood pressure, and mean arterial blood pressure in male and female WT mice (P<0.01), but the hypertensive response to Ang II was markedly attenuated in male and female PT-Nhe3-/- mice (P<0.01). Ang II impaired the pressure-natriuresis response in WT mice, whereas proximal tubule-specific deletion of NHE3 improved the pressure-natriuresis response in Ang II-infused PT-Nhe3-/- mice (P<0.01). AT1 receptor blocker losartan completely blocked Ang II-induced hypertension in both WT and PT-Nhe3-/- mice (P<0.01). However, inhibition of nitric oxide synthase with L-NG-Nitroarginine methyl ester had no effect on Ang II-induced hypertension in WT or PT-Nhe3-/- mice (not significant). Furthermore, Ang II-induced hypertension was significantly attenuated by an orally absorbable NHE3 inhibitor AVE0657. In conclusion, NHE3 in the proximal tubules of the kidney may be a therapeutical target in hypertension induced by Ang II or with increased NHE3 expression in the proximal tubules.
Collapse
Affiliation(s)
- Xiao C Li
- From the Department of Pharmacology and Toxicology (X.C.L., D.Z., X.C., X. Zheng, C.Z., J.Z., J.L.Z.), University of Mississippi Medical Center, Jackson
- Division of Nephrology, Department of Medicine (X.C.L., J.L.Z.), University of Mississippi Medical Center, Jackson
| | - Dongmin Zhu
- From the Department of Pharmacology and Toxicology (X.C.L., D.Z., X.C., X. Zheng, C.Z., J.Z., J.L.Z.), University of Mississippi Medical Center, Jackson
- Department of Anesthesiology, Shenzhen Far East Obstetrics and Gynecology Hospital, China (D.Z.)
| | - Xu Chen
- From the Department of Pharmacology and Toxicology (X.C.L., D.Z., X.C., X. Zheng, C.Z., J.Z., J.L.Z.), University of Mississippi Medical Center, Jackson
| | - Xiaowen Zheng
- From the Department of Pharmacology and Toxicology (X.C.L., D.Z., X.C., X. Zheng, C.Z., J.Z., J.L.Z.), University of Mississippi Medical Center, Jackson
- Department of Emergency Medicine, Second Affiliated Hospital, Guangxi Medical University, Nanning, China (X. Zheng, C.Z., J.Z.)
| | - Chunling Zhao
- From the Department of Pharmacology and Toxicology (X.C.L., D.Z., X.C., X. Zheng, C.Z., J.Z., J.L.Z.), University of Mississippi Medical Center, Jackson
- Department of Emergency Medicine, Second Affiliated Hospital, Guangxi Medical University, Nanning, China (X. Zheng, C.Z., J.Z.)
| | - Jianfeng Zhang
- From the Department of Pharmacology and Toxicology (X.C.L., D.Z., X.C., X. Zheng, C.Z., J.Z., J.L.Z.), University of Mississippi Medical Center, Jackson
- Department of Emergency Medicine, Second Affiliated Hospital, Guangxi Medical University, Nanning, China (X. Zheng, C.Z., J.Z.)
| | - Manoocher Soleimani
- Division of Nephrology and Hypertension, Department of Internal Medicine, The University of Cincinnati College of Medicine, OH (M.S.)
| | - Isabelle Rubera
- Laboratoire de Physiomédecine Moléculaire, LP2M, UMR-CNRS 7370, Université Côte d'Azur, Nice Cedex 2, France (I.R., M.T.)
| | - Michel Tauc
- Laboratoire de Physiomédecine Moléculaire, LP2M, UMR-CNRS 7370, Université Côte d'Azur, Nice Cedex 2, France (I.R., M.T.)
| | - Xinchun Zhou
- Department of Pathology (X. Zhou), University of Mississippi Medical Center, Jackson
| | - Jia L Zhuo
- From the Department of Pharmacology and Toxicology (X.C.L., D.Z., X.C., X. Zheng, C.Z., J.Z., J.L.Z.), University of Mississippi Medical Center, Jackson
- Division of Nephrology, Department of Medicine (X.C.L., J.L.Z.), University of Mississippi Medical Center, Jackson
| |
Collapse
|
35
|
Fernandes R, Garver H, Harkema JR, Galligan JJ, Fink GD, Xu H. Sex Differences in Renal Inflammation and Injury in High-Fat Diet-Fed Dahl Salt-Sensitive Rats. Hypertension 2019; 72:e43-e52. [PMID: 30354819 DOI: 10.1161/hypertensionaha.118.11485] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We examined the impact of sex on high-fat diet (HFD)-induced renal alterations in Dahl salt-sensitive and Sprague Dawley rats. In Dahl rats, HFD (60% kcal from fat for 24-26 weeks starting at weaning) significantly and equally increased blood pressure in males and females when compared with rats fed a control diet (10% kcal from fat). Male Dahl rats on HFD exhibited progressive renal histological injury and moderately increased renal macrophage infiltration at 10 and 24 weeks of feeding when compared with males on control diet. Female Dahl rats had lower grade renal injury and less macrophage infiltration (except at 17 weeks) than males regardless of diet. Male Dahl rats on both diets showed progressively increasing numbers of renal T-cells, a pattern not observed in females. HFD per se did not significantly affect renal T-cell number. Male Dahl rats had lower renal regulatory T-cells cell ratio than females at 24 weeks. Renal macrophage and T-cell infiltrations were highly correlated to final mean arterial pressure levels in males but not in females. Sprague Dawley rats fed HFD were normotensive without significant renal injury/inflammation after 24 weeks of feeding. In summary, HFD feeding fails to increase arterial blood pressure in Sprague Dawley rats but strongly promotes hypertension in both male and female Dahl salt-sensitive rats. Only Dahl males, however, exhibited blood pressure-associated renal inflammation and injury. Maintenance of regulatory T-cells ratio may protect against hypertension-associated renal injury/inflammation but not HFD-induced hypertension.
Collapse
Affiliation(s)
- Roxanne Fernandes
- From the Department of Pharmacology and Toxicology, Michigan State University, East Lansing (R.F., H.G., J.J.G., G.D.F., H.X.)
| | - Hannah Garver
- From the Department of Pharmacology and Toxicology, Michigan State University, East Lansing (R.F., H.G., J.J.G., G.D.F., H.X.)
| | - Jack R Harkema
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing (J.R.H.)
| | - James J Galligan
- From the Department of Pharmacology and Toxicology, Michigan State University, East Lansing (R.F., H.G., J.J.G., G.D.F., H.X.).,Neuroscience Program, Michigan State University, East Lansing (J.J.G., G.D.F., H.X.)
| | - Gregory D Fink
- From the Department of Pharmacology and Toxicology, Michigan State University, East Lansing (R.F., H.G., J.J.G., G.D.F., H.X.).,Neuroscience Program, Michigan State University, East Lansing (J.J.G., G.D.F., H.X.)
| | - Hui Xu
- From the Department of Pharmacology and Toxicology, Michigan State University, East Lansing (R.F., H.G., J.J.G., G.D.F., H.X.).,Neuroscience Program, Michigan State University, East Lansing (J.J.G., G.D.F., H.X.)
| |
Collapse
|
36
|
DuPont JJ, Kenney RM, Patel AR, Jaffe IZ. Sex differences in mechanisms of arterial stiffness. Br J Pharmacol 2019; 176:4208-4225. [PMID: 30767200 DOI: 10.1111/bph.14624] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/08/2019] [Accepted: 01/21/2019] [Indexed: 12/24/2022] Open
Abstract
Arterial stiffness progressively increases with aging and is an independent predictor of cardiovascular disease (CVD) risk. Evidence supports that there are sex differences in the time course of aging-related arterial stiffness and the associated CVD risk, which increases disproportionately in postmenopausal women. The association between arterial stiffness and mortality is almost twofold higher in women versus men. The differential clinical characteristics of the development of arterial stiffness between men and women indicate the involvement of sex-specific mechanisms. This review summarizes the current literature on sex differences in vascular stiffness induced by aging, obesity, hypertension, and sex-specific risk factors as well as the impact of hormonal status, diet, and exercise on vascular stiffness in males and females. An understanding of the mechanisms driving sex differences in vascular stiffness has the potential to identify novel sex-specific therapies to lessen CVD risk, the leading cause of death in males and females. LINKED ARTICLES: This article is part of a themed section on The Importance of Sex Differences in Pharmacology Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.21/issuetoc.
Collapse
Affiliation(s)
- Jennifer J DuPont
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Rachel M Kenney
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Ayan R Patel
- Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America.,Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| |
Collapse
|
37
|
Crajoinas RO, Polidoro JZ, Girardi ACC. The potential role of myosin motor proteins in mediating the subcellular distribution of NHE3 in the renal proximal tubule. Am J Physiol Renal Physiol 2019; 316:F986-F992. [PMID: 30864843 DOI: 10.1152/ajprenal.00577.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Isoform 3 of the Na+/H+ exchanger (NHE3) is responsible for the majority of the reabsorption of NaCl, NaHCO3, and, consequently, water in the renal proximal tubule. As such, this transporter plays an essential role in acid-base balance and extracellular fluid volume homeostasis and determining systemic arterial blood pressure levels. NHE3 activity is modulated by a number of mechanisms, including the redistribution of the transporter between the body of the microvilli (where NHE3 is active) and the base of the microvilli (where NHE3 is less active). Although the physiological, pathophysiological, and pharmacological importance of the subcellular distribution of NHE3 has been well established, the exact mechanism whereby NHE3 is translocated along microvilli microdomains of the proximal tubule apical membrane is unknown. Nonmuscle myosin IIA and unconventional myosin VI move cargoes in anterograde and retrograde directions, respectively, and are known to redistribute along with NHE3 in the proximal tubule in response to a variety of natriuretic and antinatriuretic stimuli, including stimulation or inhibition of the renin-angiotensin system, high dietary Na+ intake, and high blood pressure. Therefore, this review aims to discuss the current evidence that suggests a potential role of myosin IIA and myosin VI in mediating the subcellular distribution of NHE3 along the kidney proximal tubule microvilli and their possible contribution in modifying NHE3-mediated Na+ reabsorption under both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Renato O Crajoinas
- Heart Institute (InCor), University of São Paulo Medical School , São Paulo , Brazil
| | - Juliano Z Polidoro
- Heart Institute (InCor), University of São Paulo Medical School , São Paulo , Brazil
| | - Adriana C C Girardi
- Heart Institute (InCor), University of São Paulo Medical School , São Paulo , Brazil
| |
Collapse
|
38
|
Faulkner JL, Harwood D, Bender L, Shrestha L, Brands MW, Morwitzer MJ, Kennard S, Antonova G, de Chantemèle EJB. Lack of Suppression of Aldosterone Production Leads to Salt-Sensitive Hypertension in Female but Not Male Balb/C Mice. Hypertension 2018; 72:1397-1406. [PMID: 30571230 PMCID: PMC6309424 DOI: 10.1161/hypertensionaha.118.11303] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/11/2018] [Indexed: 12/26/2022]
Abstract
Clinical studies indicate that salt-sensitive hypertension is more prevalent in women than in men. However, animal models of salt sensitivity have primarily focused on the mechanisms of salt sensitivity in male animals; therefore, elucidation of these mechanisms in female animal models is needed. We have previously shown that female Balb/C mice have higher aldosterone synthase expression and aldosterone production than males. We hypothesized that female Balb/C mice develop salt-sensitive increases in blood pressure. Seven-day feeding of a 4% NaCl high-salt (HS) diet increased blood pressure in female mice without altering blood pressure in males. Females on an HS diet displayed no apparent increases in sodium retention as assessed by 24-hour urine collection, sodium balance measure, and saline loading excretion analysis. Females on an HS diet exhibited lower renin-angiotensin system activity (plasma Ang II [angiotensin II], plasma renin activity, and ACE [angiotensin-converting enzyme] activity) compared with males but developed a salt-induced elevation in adrenal aldosterone synthase expression and retained higher aldosterone levels than males on HS. This resulted in a higher aldosterone/plasma renin activity ratio in females compared with males on HS feeding. Adrenal mRNA expression of angiotensinogen and leptin receptor was increased in female mice on an HS diet. HS impaired endothelium-dependent relaxation in female mice only. MR (mineralocorticoid receptor) inhibition (eplerenone) restored blood pressure and endothelial function in females on an HS diet. Collectively, these data indicate that Balb/C mice develop sex-discrepant salt-sensitive hypertension likely via aldosterone-MR-mediated mechanisms involving impaired endothelium-dependent relaxation in females only. This study presents the first model of spontaneous sex-specific salt sensitivity, which mimics the human pathology.
Collapse
Affiliation(s)
- Jessica L Faulkner
- Vascular Biology Center, Medical College of Georgia at Augusta University
| | - Daisy Harwood
- Vascular Biology Center, Medical College of Georgia at Augusta University
| | - Lily Bender
- Vascular Biology Center, Medical College of Georgia at Augusta University
| | - Lenee Shrestha
- Vascular Biology Center, Medical College of Georgia at Augusta University
| | - Michael W. Brands
- Physiology Department, Medical College of Georgia at Augusta University
| | - M. Jane Morwitzer
- Vascular Biology Center, Medical College of Georgia at Augusta University
| | - Simone Kennard
- Vascular Biology Center, Medical College of Georgia at Augusta University
| | - Galina Antonova
- Vascular Biology Center, Medical College of Georgia at Augusta University
| | | |
Collapse
|
39
|
Sanz AB, Ramos AM, Soler MJ, Sanchez-Niño MD, Fernandez-Fernandez B, Perez-Gomez MV, Ortega MR, Alvarez-Llamas G, Ortiz A. Advances in understanding the role of angiotensin-regulated proteins in kidney diseases. Expert Rev Proteomics 2018; 16:77-92. [DOI: 10.1080/14789450.2018.1545577] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Ana Belén Sanz
- Nephrology, IIS-Fundacion Jimenez Diaz and Universidad Autonoma de Madrid, Madrid, Spain
| | - Adrian Mario Ramos
- Nephrology, IIS-Fundacion Jimenez Diaz and Universidad Autonoma de Madrid, Madrid, Spain
| | - Maria Jose Soler
- Department of Nephrology, Hospital del Mar-IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | | | | | | | - Marta Ruiz Ortega
- Nephrology, IIS-Fundacion Jimenez Diaz and Universidad Autonoma de Madrid, Madrid, Spain
| | - Gloria Alvarez-Llamas
- Nephrology, IIS-Fundacion Jimenez Diaz and Universidad Autonoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Nephrology, IIS-Fundacion Jimenez Diaz and Universidad Autonoma de Madrid, Madrid, Spain
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Angiotensin II is a main regulator of kidney function. Renal actions mediated by the angiotensin AT1 receptor have been well known for many years. In contrast, several details of angiotensin AT2 receptor actions in kidney physiology and pathophysiology were only described very recently. These findings are reviewed in this article. RECENT FINDINGS Regarding the role of the angiotensin AT2 receptor in kidney physiology, a major recent finding was that the AT2 receptor-mediated inhibition of Na-H exchanger-3 and Na/K-ATPase in the renal proximal tubules is caused by internalisation of these transporters, thus reducing reabsorption and increasing natriuresis/diuresis. Regarding renal pathology, several studies demonstrated an attenuation of renal injury caused by diabetes or by obesity with or without high-salt diet through anti-inflammatory, antifibrotic, and antioxidative mechanisms. Generally, AT2 receptor expression seems increased and AT2 receptor-mediated effects stronger in female and obese animals. SUMMARY The recent findings about the role of the angiotensin AT2 receptor in renal health and disease strongly suggest that pharmacological targeting of this receptor with selective agonists is a promising therapeutic strategy for inducing diuresis/natriuresis (also additive to established diuretics) and for the treatment of diabetic nephropathy or kidney disease of other pathogenesis.
Collapse
|
41
|
Frame AA, Wainford RD. Mechanisms of altered renal sodium handling in age-related hypertension. Am J Physiol Renal Physiol 2018; 315:F1-F6. [PMID: 29442548 DOI: 10.1152/ajprenal.00594.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The prevalence of hypertension rises with age to approximately two out of three adults over the age of 60 in the United States. Although the mechanisms underlying age-related hypertension are incompletely understood, sodium homeostasis is critical to the long-term regulation of blood pressure and there is strong evidence that aging is associated with alterations in renal sodium handling. This minireview focuses on recent advancements in our understanding of the vascular, neurohumoral, and renal mechanisms that influence sodium homeostasis and promote age-related hypertension.
Collapse
Affiliation(s)
- Alissa A Frame
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine and The Whitaker Cardiovascular Institute , Boston, Massachusetts
| | - Richard D Wainford
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine and The Whitaker Cardiovascular Institute , Boston, Massachusetts
| |
Collapse
|
42
|
Colafella KMM, Denton KM. Sex-specific differences in hypertension and associated cardiovascular disease. Nat Rev Nephrol 2018; 14:185-201. [PMID: 29380817 DOI: 10.1038/nrneph.2017.189] [Citation(s) in RCA: 314] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although intrinsic mechanisms that regulate arterial blood pressure (BP) are similar in men and women, marked variations exist at the molecular, cellular and tissue levels. These physiological disparities between the sexes likely contribute to differences in disease onset, susceptibility, prevalence and treatment responses. Key systems that are important in the development of hypertension and cardiovascular disease (CVD), including the sympathetic nervous system, the renin-angiotensin-aldosterone system and the immune system, are differentially activated in males and females. Biological age also contributes to sexual dimorphism, as premenopausal women experience a higher degree of cardioprotection than men of similar age. Furthermore, sex hormones such as oestrogen and testosterone as well as sex chromosome complement likely contribute to sex differences in BP and CVD. At the cellular level, differences in cell senescence pathways may contribute to increased longevity in women and may also limit organ damage caused by hypertension. In addition, many lifestyle and environmental factors - such as smoking, alcohol consumption and diet - may influence BP and CVD in a sex-specific manner. Evidence suggests that cardioprotection in women is lost under conditions of obesity and type 2 diabetes mellitus. Treatment strategies for hypertension and CVD that are tailored according to sex could lead to improved outcomes for affected patients.
Collapse
Affiliation(s)
- Katrina M Mirabito Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University Wellington Road, Clayton, Victoria 3800, Australia.,Department of Physiology, Monash University, 26 Innovation Walk, Clayton, Victoria 3800, Australia.,Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, Netherlands
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University Wellington Road, Clayton, Victoria 3800, Australia.,Department of Physiology, Monash University, 26 Innovation Walk, Clayton, Victoria 3800, Australia
| |
Collapse
|
43
|
Veiras LC, Girardi ACC, Curry J, Pei L, Ralph DL, Tran A, Castelo-Branco RC, Pastor-Soler N, Arranz CT, Yu ASL, McDonough AA. Sexual Dimorphic Pattern of Renal Transporters and Electrolyte Homeostasis. J Am Soc Nephrol 2017; 28:3504-3517. [PMID: 28774999 PMCID: PMC5698077 DOI: 10.1681/asn.2017030295] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/20/2017] [Indexed: 12/30/2022] Open
Abstract
Compared with males, females have lower BP before age 60, blunted hypertensive response to angiotensin II, and a leftward shift in pressure natriuresis. This study tested the concept that this female advantage associates with a distinct sexual dimorphic pattern of transporters along the nephron. We applied quantitative immunoblotting to generate profiles of transporters, channels, claudins, and selected regulators in both sexes and assessed the physiologic consequences of the differences. In rats, females excreted a saline load more rapidly than males did. Compared with the proximal tubule of males, the proximal tubule of females had greater phosphorylation of Na+/H+ exchanger isoform 3 (NHE3), distribution of NHE3 at the base of the microvilli, and less abundant expression of Na+/Pi cotransporter 2, claudin-2, and aquaporin 1. These changes associated with less bicarbonate reabsorption and higher lithium clearance in females. The distal nephrons of females had a higher abundance of total and phosphorylated Na+/Cl- cotransporter (NCC), claudin-7, and cleaved forms of epithelial Na+ channel (ENaC) α and γ subunits, which associated with a lower baseline plasma K+ concentration. A K+-rich meal increased the urinary K+ concentration and decreased the level of renal phosphorylated NCC in females. Notably, we observed similar abundance profiles in female versus male C57BL/6 mice. These results define sexual dimorphic phenotypes along the nephron and suggest that lower proximal reabsorption in female rats expedites excretion of a saline load and enhances NCC and ENaC abundance and activation, which may facilitate K+ secretion and set plasma K+ at a lower level.
Collapse
Affiliation(s)
| | | | - Joshua Curry
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Lei Pei
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | | | - An Tran
- Department of Integrative Anatomical Sciences and
| | - Regiane C Castelo-Branco
- Department of Physiology and Biophysics, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil; and
| | - Nuria Pastor-Soler
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Cristina T Arranz
- University of Buenos Aires, National Council of Science and Technology, Buenos Aires, Argentina
| | - Alan S L Yu
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | | |
Collapse
|
44
|
Sullivan JC, Gillis EE. Sex and gender differences in hypertensive kidney injury. Am J Physiol Renal Physiol 2017; 313:F1009-F1017. [PMID: 28724606 PMCID: PMC5668592 DOI: 10.1152/ajprenal.00206.2017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/11/2017] [Accepted: 07/12/2017] [Indexed: 12/30/2022] Open
Abstract
Hypertension is a complex, multifaceted disorder, affecting ~1 in 3 adults in the United States. Although hypertension occurs in both men and women, there are distinct sex differences in the way in which they develop hypertension, with women having a lower incidence of hypertension until the sixth decade of life. Despite observed sex differences in hypertension, little is known about the molecular mechanisms underlying the development of hypertension in females, primarily because of their underrepresentation in both clinical and experimental animal studies. The first goal of this review is to provide a concise overview of the participation of women in clinical trials, including a discussion of the importance of including females in basic science research, as recently mandated by the National Institutes of Health. The remaining portion of the review is dedicated to identifying clinical and experimental animal studies that concentrate on gender and sex differences in hypertensive kidney disease, ending with a proposed role for T cells in mediating sex differences in blood pressure.
Collapse
Affiliation(s)
| | - Ellen E Gillis
- Department of Physiology, Augusta University, Augusta, Georgia
| |
Collapse
|
45
|
Sandberg K, Pai AV, Maddox T. Sex and rigor: the TGF-β blood pressure affair. Am J Physiol Renal Physiol 2017; 313:F1087-F1088. [PMID: 28794068 DOI: 10.1152/ajprenal.00381.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Kathryn Sandberg
- Division of Nephrology and Hypertension, Department of Medicine, Georgetown University, Washington, DC; and .,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC
| | - Amrita V Pai
- Division of Nephrology and Hypertension, Department of Medicine, Georgetown University, Washington, DC; and.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC
| | - Taylor Maddox
- Division of Nephrology and Hypertension, Department of Medicine, Georgetown University, Washington, DC; and
| |
Collapse
|
46
|
Wang Z, Zhu Q, Wang W, Yi F, Li PL, Boini KM, Li N. Infusion of Valproic Acid Into the Renal Medulla Activates Stem Cell Population and Attenuates Salt-Sensitive Hypertension in Dahl S Rats. Cell Physiol Biochem 2017; 42:1264-1273. [PMID: 28693025 DOI: 10.1159/000478955] [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: 08/02/2016] [Accepted: 04/25/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Our previous study has detected a stem cell deficiency in the renal medulla in Dahl salt-sensitive (S) rats. This study determined whether infusion of valproic acid (VA), an agent known to stimulate the stem cell function, attenuated salt-sensitive hypertension in Dahl S rats. METHODS Uninephrectomized Dahl S rats were infused with vehicle or VA (50mg/kg/d) into the renal medulla and fed with a low (LS) or high salt diet (HS). Stem cell marker and number were analyzed by immunohistochemistry, Real-time RT-PCR and Western blot. Sodium excretion and blood pressure were measured. RESULTS VA significantly increased the mRNA and protein levels of FGF2, a stem cell niche factor, and CD133, a stem cell marker. The number of CD133+ cells was significantly increased in the renal medulla in VA-treated rats. Meanwhile, high salt-induced increases in the mRNA level of proinflammatory factors interleukin-1β and interleukin-6 were blocked in VA-treated rats. Functionally, sodium excretion in response to the blood pressure increase and acute sodium loading was significantly enhanced, sodium retention attenuated, high salt-induced increase of blood pressure reduced in VA-treated rats. CONCLUSION Activation of stem cell function by VA inhibits the activation of proinflammatory factors and attenuates salt-sensitive hypertension in Dahl S rats.
Collapse
Affiliation(s)
- Zhengchao Wang
- Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, China.,Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Qing Zhu
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA.,Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Weili Wang
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Fan Yi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Pin-Lan Li
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Krishna M Boini
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Ningjun Li
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
47
|
Samuel CS, Royce SG, Hewitson TD, Denton KM, Cooney TE, Bennett RG. Anti-fibrotic actions of relaxin. Br J Pharmacol 2017; 174:962-976. [PMID: 27250825 PMCID: PMC5406285 DOI: 10.1111/bph.13529] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/19/2016] [Accepted: 05/23/2016] [Indexed: 12/19/2022] Open
Abstract
Fibrosis refers to the hardening or scarring of tissues that usually results from aberrant wound healing in response to organ injury, and its manifestations in various organs have collectively been estimated to contribute to around 45-50% of deaths in the Western world. Despite this, there is currently no effective cure for the tissue structural and functional damage induced by fibrosis-related disorders. Relaxin meets several criteria of an effective anti-fibrotic based on its specific ability to inhibit pro-fibrotic cytokine and/or growth factor-mediated, but not normal/unstimulated, fibroblast proliferation, differentiation and matrix production. Furthermore, relaxin augments matrix degradation through its ability to up-regulate the release and activation of various matrix-degrading matrix metalloproteinases and/or being able to down-regulate tissue inhibitor of metalloproteinase activity. Relaxin can also indirectly suppress fibrosis through its other well-known (anti-inflammatory, antioxidant, anti-hypertrophic, anti-apoptotic, angiogenic, wound healing and vasodilator) properties. This review will outline the organ-specific and general anti-fibrotic significance of exogenously administered relaxin and its mechanisms of action that have been documented in various non-reproductive organs such as the cardiovascular system, kidney, lung, liver, skin and tendons. In addition, it will outline the influence of sex on relaxin's anti-fibrotic actions, highlighting its potential as an emerging anti-fibrotic therapeutic. LINKED ARTICLES This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc.
Collapse
Affiliation(s)
- C S Samuel
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of PharmacologyMonash UniversityMelbourneVic.Australia
| | - S G Royce
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of PharmacologyMonash UniversityMelbourneVic.Australia
| | - T D Hewitson
- Department of NephrologyRoyal Melbourne HospitalMelbourneVic.Australia
| | - K M Denton
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of PhysiologyMonash UniversityMelbourneVic.Australia
| | - T E Cooney
- University of Pittsburgh Medical Centre (UPMC) HamotEriePAUSA
| | - R G Bennett
- Research Service 151VA Nebraska‐Western Iowa Health Care SystemOmahaNEUSA
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNEUSA
| |
Collapse
|
48
|
Epochs in the depressor/pressor balance of the renin-angiotensin system. Clin Sci (Lond) 2017; 130:761-71. [PMID: 27128801 DOI: 10.1042/cs20150939] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/01/2016] [Indexed: 12/17/2022]
Abstract
The renin-angiotensin system (RAS) plays a commanding role in the regulation of extracellular fluid homoeostasis. Tigerstadt and Bergman first identified the RAS more than two centuries ago. By the 1980s a voyage of research and discovery into the mechanisms and actions of this system led to the development of drugs that block the RAS, which have become the mainstay for the treatment of cardiovascular and renal disease. In the last 25 years new components of the RAS have come to light, including the angiotensin type 2 receptor (AT2R) and the angiotensin-converting enzyme 2 (ACE2)/angiotensin-(1-7) [Ang(1-7)]/Mas receptor (MasR) axis. These have been shown to counter the classical actions of angiotensin II (AngII) at the predominant angiotensin type 1 receptor (AT1R). Our studies, and those of others, have demonstrated that targeting these depressor RAS pathways may be therapeutically beneficial. It is apparent that the evolution of both the pressor and depressor RAS pathways is distinct throughout life and that the depressor/pressor balance of the RAS vary between the sexes. These temporal patterns of expression suggest that therapies targeting the RAS could be optimized for discrete epochs in life.
Collapse
|
49
|
Carey RM. AT2 Receptors: Potential Therapeutic Targets for Hypertension. Am J Hypertens 2017; 30:339-347. [PMID: 27664954 DOI: 10.1093/ajh/hpw121] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 09/07/2016] [Indexed: 12/15/2022] Open
Abstract
The renin-angiotensin system (RAS) is arguably the most important and best studied hormonal system in the control of blood pressure (BP) and the pathogenesis of hypertension. The RAS features its main effector angiotensin II (Ang II) acting via its 2 major receptors, angiotensin type-1(AT1R) and type-2 (AT2R). In general, AT2Rs oppose the detrimental actions of Ang II via AT1Rs. AT2R activation induces vasodilation and natriuresis, but its effects to lower BP in hypertension have not been as clear as anticipated. Recent studies, however, have demonstrated that acute and chronic AT2R stimulation can induce natriuresis and lower BP in the Ang II infusion model of experimental hypertension. AT2R activation induces receptor recruitment from intracellular sites to the apical plasma membranes of renal proximal tubule cells via a bradykinin, nitric oxide, and cyclic guanosine 3',5' monophosphate signaling pathway that results in internalization and inactivation of sodium (Na+) transporters Na+-H+ exchanger-3 and Na+/K+ATPase. These responses do not require the presence of concurrent AT1R blockade and are effective both in the prevention and reversal of hypertension. This review will address the role of AT2Rs in the control of BP and Na+ excretion and the case for these receptors as potential therapeutic targets for hypertension in humans.
Collapse
Affiliation(s)
- Robert M Carey
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia, USA
| |
Collapse
|
50
|
Barsha G, Denton KM, Mirabito Colafella KM. Sex- and age-related differences in arterial pressure and albuminuria in mice. Biol Sex Differ 2016; 7:57. [PMID: 27895890 PMCID: PMC5109725 DOI: 10.1186/s13293-016-0110-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/22/2016] [Indexed: 11/25/2022] Open
Abstract
Background Animal models have become valuable experimental tools for understanding the pathophysiology and therapeutic interventions in cardiovascular disease. Yet to date, few studies document the age- and sex-related differences in arterial pressure, circadian rhythm, and renal function in normotensive mice under basal conditions, across the life span. We hypothesized that mice display similar sex- and age-related differences in arterial pressure and renal function to humans. Methods Mean arterial pressure (MAP) and circadian rhythm of arterial pressure were measured over 3 days via radiotelemetry, in 3- and 5-month-old (adult) and 14- and 18-month-old (aged) FVB/N and in 5-month-old (adult) C57BL/6 male and female normotensive mice. In FVB/N mice, albuminuria from 24-h urine samples as well as body, heart, and kidney weights were measured at each age. Results Twenty-four-hour MAP was greater in males than females at 3, 5, and 14 months of age. A similar sex difference in arterial pressure was observed in C57BL/6 mice at 5 months of age. In FVB/N mice, 24-h MAP increased with age, with females displaying a greater increase between 3 and 18 months of age than males, such that MAP was no longer different between the sexes at 18 months of age. A circadian pattern was observed in arterial pressure, heart rate, and locomotor activity, with values for each greater during the active (night/dark) than the inactive (day/light) period. The night-day dip in MAP was greater in males and increased with age in both sexes. Albuminuria was greater in males than females, increased with age in both sexes, and rose to a greater level in males than females at 18 months of age. Conclusions Arterial pressure and albuminuria increase in an age- and sex-specific manner in mice, similar to patterns observed in humans. Thus, mice represent a useful model for studying age and sex differences in the regulation of arterial pressure and renal disease. Understanding the mechanisms that underlie the pathophysiology of cardiovascular disease may lead to new and better-tailored therapies for men and women.
Collapse
Affiliation(s)
- Giannie Barsha
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Clayton, Australia.,Department of Physiology, Monash University, 26 Innovation Walk (Building 13F), Clayton, VIC 3800 Australia
| | - Kate M Denton
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Clayton, Australia.,Department of Physiology, Monash University, 26 Innovation Walk (Building 13F), Clayton, VIC 3800 Australia
| | - Katrina M Mirabito Colafella
- Cardiovascular Program, Monash Biomedicine Discovery Institute, Clayton, Australia.,Department of Physiology, Monash University, 26 Innovation Walk (Building 13F), Clayton, VIC 3800 Australia
| |
Collapse
|