1
|
Saleem M, Aden LA, Mutchler AL, Basu C, Ertuglu LA, Sheng Q, Penner N, Hemnes AR, Park JH, Ishimwe JA, Laffer CL, Elijovich F, Wanjalla CN, de la Visitacion N, Kastner PD, Albritton CF, Ahmad T, Haynes AP, Yu J, Graber MK, Yasmin S, Wagner KU, Sayeski PP, Hatzopoulos AK, Gamazon ER, Bick AG, Kleyman TR, Kirabo A. Myeloid-Specific JAK2 Contributes to Inflammation and Salt Sensitivity of Blood Pressure. Circ Res 2024; 135:890-909. [PMID: 39263750 PMCID: PMC11466692 DOI: 10.1161/circresaha.124.323595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Salt sensitivity of blood pressure (SSBP), characterized by acute changes in blood pressure with changes in dietary sodium intake, is an independent risk factor for cardiovascular disease and mortality in people with and without hypertension. We previously found that elevated sodium concentration activates antigen-presenting cells (APCs), resulting in high blood pressure, but the mechanisms are unknown. Here, we hypothesized that APC-specific JAK2 (Janus kinase 2) through STAT3 (signal transducer and activator of transcription 3) and SMAD3 (small mothers against decapentaplegic homolog 3) contributes to SSBP. METHODS We performed bulk or single-cell transcriptomic analyses following in vitro monocytes exposed to high salt and in vivo high sodium treatment in humans using a rigorous salt-loading/depletion protocol to phenotype SSBP. We also used a myeloid cell-specific CD11c+ JAK2 knockout mouse model and measured blood pressure with radiotelemetry after N-omega-nitro-L-arginine-methyl ester and a high salt diet treatment. We used flow cytometry for immunophenotyping and measuring cytokine levels. Fluorescence in situ hybridization and immunohistochemistry were performed to spatially visualize the kidney's immune cells and cytokine levels. Echocardiography was performed to assess cardiac function. RESULTS We found that high salt treatment upregulates gene expression of the JAK/STAT/SMAD pathway while downregulating inhibitors of this pathway, such as suppression of cytokine signaling and cytokine-inducible SH2, in human monocytes. Expression of the JAK2 pathway genes mirrored changes in blood pressure after salt loading and depletion in salt-sensitive but not salt-resistant humans. Ablation of JAK2, specifically in CD11c+ APCs, attenuated salt-induced hypertension in mice with SSBP. Mechanistically, we found that SMAD3 acted downstream of JAK2 and STAT3, leading to increased production of highly reactive isolevuglandins and proinflammatory cytokine IL (interleukin)-6 in renal APCs, which activate T cells and increase production of IL-17A, IL-6, and TNF-α (tumor necrosis factor-alpha). CONCLUSIONS Our findings reveal the APC JAK2 signaling pathway as a potential target for the diagnosis and treatment of SSBP in humans.
Collapse
Affiliation(s)
- Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Luul A Aden
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Ashley L Mutchler
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Chitra Basu
- Department of Medicine, Division of Genetic Medicine (C.B., E.R.G.), Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Division of Cardiovascular Medicine (C.B., A.K.H.), Vanderbilt University Medical Center, Nashville, TN
| | - Lale A Ertuglu
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Quanhu Sheng
- Department of Biostatistics (Q.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Niki Penner
- Division of Allergy, Pulmonary, and Critical Care Medicine (N.P., A.R.H.)
| | - Anna R Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine (N.P., A.R.H.)
| | - Jennifer H Park
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Jeanne A Ishimwe
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Cheryl L Laffer
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | | | - Celestine N Wanjalla
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Nestor de la Visitacion
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Paul D Kastner
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Claude F Albritton
- School of Graduate Studies, Meharry Medical College, Nashville, TN (C.F.A.)
| | - Taseer Ahmad
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Punjab, Pakistan (T.A.)
| | - Alexandria P Haynes
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Justin Yu
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Meghan K Graber
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Sharia Yasmin
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Kay-Uwe Wagner
- Wayne State University, Department of Oncology and Tumor Biology Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI (K.-U.W.)
| | - Peter P Sayeski
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville (P.P.S.)
| | - Antonis K Hatzopoulos
- Department of Medicine, Division of Cardiovascular Medicine (C.B., A.K.H.), Vanderbilt University Medical Center, Nashville, TN
| | - Eric R Gamazon
- Department of Medicine, Division of Genetic Medicine (C.B., E.R.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Alexander G Bick
- Division of Genetic Medicine (A.G.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, PA (T.R.K.)
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation (A.K.)
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN (A.K.)
| |
Collapse
|
2
|
Sabouri M, Zheng X, Irwin BJ, Machin DR. Effects of excess sodium consumption on arterial function in C57BL/6 mice. Am J Physiol Heart Circ Physiol 2024; 327:H896-H907. [PMID: 39150393 PMCID: PMC11482244 DOI: 10.1152/ajpheart.00242.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Excess sodium consumption contributes to arterial dysfunction in humans. The C57BL/6 strain of mice has been used to identify mechanisms by which arterial dysfunction occurs after excess sodium consumption. However, there are concerns that C57BL/6 mice have strain-specific resistance to high-sodium (HS) diet-induced hypertension. To address this concern, we performed a meta-analysis to determine if excess sodium consumption in C57BL/6 mice induces arterial dysfunction. Databases were searched for HS versus standard diet studies that measured arterial function [i.e., systolic blood pressure (BP), endothelium-dependent dilation (EDD), and central arterial stiffness] in C57BL/6 mice. A total of 39 studies were included, demonstrating that the HS condition resulted in higher systolic BP than control mice with a mean difference of 9.8 mmHg (95% confidence interval [CI] = [5.6, 14], P < 0.001). Subgroup analysis indicated that the systolic BP was higher in HS compared with the control condition when measured during night compared with daytime with telemetry (P < 0.001). We also identified that the difference in systolic BP between HS and control was ∼2.5-fold higher when administered through drinking water than through food (P < 0.001). A total of 12 studies were included, demonstrating that the HS condition resulted in lower EDD than control with a weighted mean difference of -12.0% (95% CI = [-20.0, -4.1], P = 0.003). It should be noted that there was considerable variability across studies with more than half of the studies showing no effect of the HS condition on systolic BP or EDD. In summary, excess sodium consumption elevates systolic BP and impairs EDD in C57BL/6 mice.NEW & NOTEWORTHY C57BL/6 mice are perceived as resistant to high-sodium diet-induced arterial dysfunction. This meta-analysis demonstrates that excess sodium consumption elevates blood pressure and impairs endothelium-dependent dilation in C57BL/6 mice. Nighttime measurements show more pronounced blood pressure elevation. In addition, sodium administration via drinking water, compared with food, induces a greater blood pressure elevation. These findings may be influenced by outlier studies, as the majority of studies showed no adverse effect of excess sodium consumption on arterial function.
Collapse
Affiliation(s)
- Mostafa Sabouri
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, United States
| | - Xiangyu Zheng
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, United States
| | - Bryan J Irwin
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States
| | - Daniel R Machin
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
3
|
Verouti S, Aeschlimann G, Wang Q, Del Olmo DA, Peyter AC, Menétrey S, Winter DV, Odermatt A, Pearce D, Hummler E, Vanderriele PE. Salt-sensitive hypertension in GR mutant rats is associated with altered plasma polyunsaturated fatty acid levels and aortic vascular reactivity. Pflugers Arch 2024:10.1007/s00424-024-03014-y. [PMID: 39256246 DOI: 10.1007/s00424-024-03014-y] [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: 04/30/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024]
Abstract
In humans, glucocorticoid resistance is attributed to mutations in the glucocorticoid receptor (GR). Most of these mutations result in decreased ligand binding, transactivation, and/or translocation, albeit with normal protein abundances. However, there is no clear genotype‒phenotype relationship between the severity or age at disease presentation and the degree of functional loss of the receptor. Previously, we documented that a GR+/- rat line developed clinical features of glucocorticoid resistance, namely, hypercortisolemia, adrenal hyperplasia, and salt-sensitive hypertension. In this study, we analyzed the GR+/em4 rat model heterozygously mutant for the deletion of exon 3, which encompasses the second zinc finger, including the domains of DNA binding, dimerization, and nuclear localization signals. On a standard diet, mutant rats exhibited a trend toward increased corticosterone levels and a normal systolic blood pressure and heart rate but presented with adrenal hyperplasia. They exhibited increased adrenal soluble epoxide hydroxylase (sEH), favoring an increase in less active polyunsaturated fatty acids. Indeed, a significant increase in nonactive omega-3 and omega-6 polyunsaturated fatty acids, such as 5(6)-DiHETrE or 9(10)-DiHOME, was observed with advanced age (10 versus 5 weeks old) and following a switch to a high-salt diet accompanied by salt-sensitive hypertension. In thoracic aortas, a reduced soluble epoxide hydrolase (sEH) protein abundance resulted in altered vascular reactivity upon a standard diet, which was blunted upon a high-salt diet. In conclusion, mutations in the GR affecting the ligand-binding domain as well as the dimerization domain resulted in deregulated GR signaling, favoring salt-sensitive hypertension in the absence of obvious mineralocorticoid excess.
Collapse
Affiliation(s)
- S Verouti
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- National Center of Competence in Research, Kidney.CH, Lausanne, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - G Aeschlimann
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Q Wang
- Division of Nephrology and Hypertension, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - D Ancin Del Olmo
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - A C Peyter
- Neonatal Research Laboratory, Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - S Menétrey
- Neonatal Research Laboratory, Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - D V Winter
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - A Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - D Pearce
- Department of Medicine and Cellular & Molecular Pharmacology, University of California, San Francisco, USA
| | - E Hummler
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- National Center of Competence in Research, Kidney.CH, Lausanne, Switzerland
| | - P E Vanderriele
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
- National Center of Competence in Research, Kidney.CH, Lausanne, Switzerland.
| |
Collapse
|
4
|
Faienza S, Citterio L, Messaggio E, Zagato L, Lanzani C, Simonini M, Canciani B, Sanvito F, Rampoldi L, Pavlovic D, Manunta P. A novel mouse model recapitulates the effects of rs2254524 variant in the lanosterol synthase gene on salt sensitivity and organ damage. J Hypertens 2024:00004872-990000000-00523. [PMID: 39248148 DOI: 10.1097/hjh.0000000000003843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024]
Abstract
OBJECTIVE The blood pressure (BP) response to salt intake (salt sensitivity) shows great variability among individuals and is more frequent in hypertensive patients. Elevated levels of the steroid hormone Endogenous Ouabain (EO) are associated with hypertension (HT) and salt sensitivity. The lanosterol synthase gene ( LSS ) plays a key role in the biosynthesis of steroids and its rs2254524 variant (Val642Leu) is linked to salt sensitivity in humans. This study aims to investigate the pathophysiological significance of the Lss missense variation in a new knock-in mouse model of salt-sensitive HT onset. METHODS We generated a mouse model carrying the murine homolog (Val643Leu) of the human LSS variant. C57BL/6N LssV643L/V643L mice were fed different NaCl diets (low-salt, LSD; normal-salt, NSD; high-salt, HSD) and were characterized at functional, histological, and molecular levels. RESULTS At baseline, mutant mice showed an enlarged kidney compared to the wild-type (WT) counterpart, but the Lss V643L variant did not affect EO biosynthesis nor systolic BP at 3 and 12 months. In HSD, we observed an increased systolic BP only in 12-month-old LssV643L/V643L mice, compared to NSD. Moreover, only the HSD LssV643L/V643L mice showed cardiac hypertrophy and a higher incidence of cardiac fibrosis compared to WT at 12 months. Finally, the Lss mRNA level was differentially regulated by HSD in the adrenal gland, liver, and heart of LssV643L/V643L mice compared to WT. CONCLUSIONS The novel Lss mouse model resembles the salt-sensitive HT phenotype observed in hypertensive patients and provides a good model of salt-sensitive HT and HT-mediated organ damage.
Collapse
Affiliation(s)
- Sipontina Faienza
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
- Università Vita-Salute San Raffaele
| | - Lorena Citterio
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
| | | | - Laura Zagato
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
| | - Chiara Lanzani
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
- Università Vita-Salute San Raffaele
| | - Marco Simonini
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
| | | | - Francesca Sanvito
- Pathology Unit, Department of Experimental Oncology, IRCCS Ospedale San Raffaele
| | - Luca Rampoldi
- Università Vita-Salute San Raffaele
- Molecular Genetics of Renal Disorders Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Davor Pavlovic
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Wolfson Drive, Birmingham, UK
| | - Paolo Manunta
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
- Università Vita-Salute San Raffaele
| |
Collapse
|
5
|
Sesa-Ashton G, Carnagarin R, Nolde JM, Muente I, Lee R, Macefield VG, Dawood T, Sata Y, Lambert EA, Lambert GW, Walton A, Kiuchi MG, Esler MD, Schlaich MP. Salt sensitivity risk derived from nocturnal dipping and 24-h heart rate predicts long-term blood pressure reduction following renal denervation. J Hypertens 2024; 42:922-927. [PMID: 38230602 DOI: 10.1097/hjh.0000000000003655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
BACKGROUND Renal denervation (RDN) has been consistently shown in recent sham-controlled clinical trials to reduce blood pressure (BP). Salt sensitivity is a critical factor in hypertension pathogenesis, but cumbersome to assess by gold-standard methodology. Twenty-four-hour average heart rate (HR) and mean arterial pressure (MAP) dipping, taken by ambulatory blood pressure monitoring (ABPM), stratifies patients into high, moderate, and low salt sensitivity index (SSI) risk categories. OBJECTIVES We aimed to assess whether ABPM-derived SSI risk could predict the systolic blood pressure reduction at long-term follow-up in a real-world RDN patient cohort. METHODS Sixty participants had repeat ABPM as part of a renal denervation long-term follow-up. Average time since RDN was 8.9 ± 1.2 years. Based on baseline ABPM, participants were stratified into low (HR < 70 bpm and MAP dipping > 10%), moderate (HR ≥70 bpm or MAP dipping ≤ 10%), and high (HR ≥ 70 bpm and MAP dipping ≤ 10%) SSI risk groups, respectively. RESULTS One-way ANOVA indicated a significant treatment effect ( P = 0.03) between low ( n = 15), moderate ( n = 35), and high ( n = 10) SSI risk with systolic BP reduction of 9.6 ± 3.7 mmHg, 8.4 ± 3.5 mmHg, and 28.2 ± 9.6 mmHg, respectively. Baseline BP was not significantly different between SSI Risk groups ( P = 0.18). High SSI risk independently correlated with systolic BP reduction ( P = 0.02). CONCLUSIONS Our investigation indicates that SSI risk may be a simple and accessible measure for predicting the BP response to RDN. However, the influence of pharmacological therapy on these participants is an important extraneous variable requiring testing in prospective or drug naive RDN cohorts.
Collapse
Affiliation(s)
- Gianni Sesa-Ashton
- Human Neurotransmitter and Neurovascular Hypertension & Kidney Diseases Laboratories, Baker Heart and Diabetes Institute, Melbourne
- Human Autonomic Neurophysiology Laboratory, Baker Heart and Diabetes Institute
| | - Revathy Carnagarin
- Dobney Hypertension Centre, Medical School - Royal Perth Hospital Unit and RPH Research Foundation, The University of Western Australia, Perth, Western Australia
| | - Janis M Nolde
- Dobney Hypertension Centre, Medical School - Royal Perth Hospital Unit and RPH Research Foundation, The University of Western Australia, Perth, Western Australia
| | - Ida Muente
- Dobney Hypertension Centre, Medical School - Royal Perth Hospital Unit and RPH Research Foundation, The University of Western Australia, Perth, Western Australia
| | - Rebecca Lee
- Human Neurotransmitter and Neurovascular Hypertension & Kidney Diseases Laboratories, Baker Heart and Diabetes Institute, Melbourne
| | - Vaughan G Macefield
- Human Autonomic Neurophysiology Laboratory, Baker Heart and Diabetes Institute
| | - Tye Dawood
- Human Autonomic Neurophysiology Laboratory, Baker Heart and Diabetes Institute
| | - Yusuke Sata
- Human Neurotransmitter and Neurovascular Hypertension & Kidney Diseases Laboratories, Baker Heart and Diabetes Institute, Melbourne
- Department of Cardiology, Alfred Health, Melbourne, Victoria
| | - Elisabeth A Lambert
- Iverson Health Innovation Research Institute and School of Health Sciences, Swinburne University of Technology, Melbourne
| | - Gavin W Lambert
- Iverson Health Innovation Research Institute and School of Health Sciences, Swinburne University of Technology, Melbourne
| | - Antony Walton
- Department of Cardiology, Alfred Health, Melbourne, Victoria
| | - Marcio G Kiuchi
- Dobney Hypertension Centre, Medical School - Royal Perth Hospital Unit and RPH Research Foundation, The University of Western Australia, Perth, Western Australia
| | - Murray D Esler
- Human Neurotransmitter and Neurovascular Hypertension & Kidney Diseases Laboratories, Baker Heart and Diabetes Institute, Melbourne
- Department of Cardiology, Alfred Health, Melbourne, Victoria
| | - Markus P Schlaich
- Human Neurotransmitter and Neurovascular Hypertension & Kidney Diseases Laboratories, Baker Heart and Diabetes Institute, Melbourne
- Dobney Hypertension Centre, Medical School - Royal Perth Hospital Unit and RPH Research Foundation, The University of Western Australia, Perth, Western Australia
- Departments of Cardiology and Nephrology, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
6
|
Faraci FM, Scheer FA. Hypertension: Causes and Consequences of Circadian Rhythms in Blood Pressure. Circ Res 2024; 134:810-832. [PMID: 38484034 PMCID: PMC10947115 DOI: 10.1161/circresaha.124.323515] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/16/2024] [Indexed: 03/19/2024]
Abstract
Hypertension is extremely common, affecting approximately 1 in every 2 adults globally. Chronic hypertension is the leading modifiable risk factor for cardiovascular disease and premature mortality worldwide. Despite considerable efforts to define mechanisms that underlie hypertension, a potentially major component of the disease, the role of circadian biology has been relatively overlooked in both preclinical models and humans. Although the presence of daily and circadian patterns has been observed from the level of the genome to the whole organism, the functional and structural impact of biological rhythms, including mechanisms such as circadian misalignment, remains relatively poorly defined. Here, we review the impact of daily rhythms and circadian systems in regulating blood pressure and the onset, progression, and consequences of hypertension. There is an emphasis on the impact of circadian biology in relation to vascular disease and end-organ effects that, individually or in combination, contribute to complex phenotypes such as cognitive decline and the loss of cardiac and brain health. Despite effective treatment options for some individuals, control of blood pressure remains inadequate in a substantial portion of the hypertensive population. Greater insight into circadian biology may form a foundation for novel and more widely effective molecular therapies or interventions to help in the prevention, treatment, and management of hypertension and its related pathophysiology.
Collapse
Affiliation(s)
- Frank M. Faraci
- Department of Internal Medicine, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1081
- Department of Neuroscience and Pharmacology, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1081
| | - Frank A.J.L. Scheer
- Division of Sleep Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, 02115
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, Massachusetts, 02115
| |
Collapse
|
7
|
Hoenemann JN, Moestl S, Diedrich A, Mulder E, Frett T, Petrat G, Pustowalow W, Arz M, Schmitz MT, Heusser K, Lee SMC, Jordan J, Tank J, Hoffmann F. Impact of daily artificial gravity on autonomic cardiovascular control following 60-day head-down tilt bed rest. Front Cardiovasc Med 2023; 10:1250727. [PMID: 37953766 PMCID: PMC10634666 DOI: 10.3389/fcvm.2023.1250727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/04/2023] [Indexed: 11/14/2023] Open
Abstract
Impaired cardiovascular autonomic control following space flight or immobilization may limit the ability to cope with additional hemodynamic stimuli. Head-down tilt bedrest is an established terrestrial analog for space flight and offers the opportunity to test potential countermeasures for autonomic cardiovascular deconditioning. Previous studies revealed a possible benefit of daily artificial gravity on cardiovascular autonomic control following head-down tilt bedrest, but there is a need for efficiency in a long-term study before an artificial gravity facility would be brought to space. We hypothesized that artificial gravity through short-arm centrifugation attenuates functional adaptions of autonomic function during head-down tilt bed rest. 24 healthy persons (8 women, 33.4 ± 9.3 years, 24.3 ± 2.1 kg/m2) participated in the 60-day head-down tilt bed rest (AGBRESA) study. They were assigned to three groups, 30 min/day continuous, or 6(5 min intermittent short-arm centrifugation, or a control group. We assessed autonomic cardiovascular control in the supine position and in 5 minutes 80° head-up tilt position before and immediately after bed rest. We computed heart rate variability (HRV) in the time (rmssd) and frequency domain, blood pressure variability, and baroreflex sensitivity (BRS). RR interval corrected rmssd was reduced supine (p = 0.0358) and during HUT (p = 0.0161). Heart rate variability in the high-frequency band (hf-RRI; p = 0.0004) and BRS (p < 0.0001) decreased, whereas blood pressure variability in the low-frequency band (lf-SBP, p = 0.0008) increased following bedrest in all groups. We did not detect significant interactions between bedrest and interventions. We conclude that up to daily 30 min of artificial gravity on a short-arm centrifuge with 1Gz at the center of mass do not suffice to prevent changes in autonomic cardiovascular control following 60-day of 6° head-down tilt bed rest. Clinical Trial Registration: https://drks.de/search/en/trial/DRKS00015677, identifier, DRKS00015677.
Collapse
Affiliation(s)
- J.-N. Hoenemann
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
- Department of Internal Medicine III, Division of Cardiology, Pneumology, Angiology, and Intensive Care, University of Cologne, Cologne, Germany
| | - S. Moestl
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - A. Diedrich
- Department of Medicine, Division of Clinical Pharmacology, Autonomic Dysfunction Service, Vanderbilt University, Nashville, TN, United States
| | - E. Mulder
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - T. Frett
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - G. Petrat
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - W. Pustowalow
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - M. Arz
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - M.-T. Schmitz
- Institute of Medical Biometry, Informatics and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| | - K. Heusser
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - S. M. C. Lee
- Wyle Laboratories, Life Sciences and Systems Division, Houston, TX, United States
| | - J. Jordan
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
- Head of Aerospace Medicine, University of Cologne, Germany, Cologne
| | - J. Tank
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - F. Hoffmann
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
- Department of Internal Medicine III, Division of Cardiology, Pneumology, Angiology, and Intensive Care, University of Cologne, Cologne, Germany
| |
Collapse
|
8
|
Costello HM, Krilis G, Grenier C, Severs D, Czopek A, Ivy JR, Nixon M, Holmes MC, Livingstone DEW, Hoorn EJ, Dhaun N, Bailey MA. High salt intake activates the hypothalamic-pituitary-adrenal axis, amplifies the stress response, and alters tissue glucocorticoid exposure in mice. Cardiovasc Res 2023; 119:1740-1750. [PMID: 36368681 PMCID: PMC10325699 DOI: 10.1093/cvr/cvac160] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 09/09/2022] [Accepted: 09/24/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS High salt intake is common and contributes to poor cardiovascular health. Urinary sodium excretion correlates directly with glucocorticoid excretion in humans and experimental animals. We hypothesized that high salt intake activates the hypothalamic-pituitary-adrenal axis activation and leads to sustained glucocorticoid excess. METHODS AND RESULTS In male C57BL/6 mice, high salt intake for 2-8 weeks caused an increase in diurnal peak levels of plasma corticosterone. After 2 weeks, high salt increased Crh and Pomc mRNA abundance in the hypothalamus and anterior pituitary, consistent with basal hypothalamic-pituitary-adrenal axis activation. Additionally, high salt intake amplified glucocorticoid response to restraint stress, indicative of enhanced axis sensitivity. The binding capacity of Corticosteroid-Binding Globulin was reduced and its encoding mRNA downregulated in the liver. In the hippocampus and anterior pituitary, Fkbp5 mRNA levels were increased, indicating increased glucocorticoid exposure. The mRNA expression of the glucocorticoid-regenerating enzyme, 11β-hydroxysteroid dehydrogenase Type 1, was increased in these brain areas and in the liver. Sustained high salt intake activated a water conservation response by the kidney, increasing plasma levels of the vasopressin surrogate, copeptin. Increased mRNA abundance of Tonebp and Avpr1b in the anterior pituitary suggested that vasopressin signalling contributes to hypothalamic-pituitary-adrenal axis activation by high salt diet. CONCLUSION Chronic high salt intake amplifies basal and stress-induced glucocorticoid levels and resets glucocorticoid biology centrally, peripherally and within cells.
Collapse
Affiliation(s)
- Hannah M Costello
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The Universtiy of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Georgios Krilis
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The Universtiy of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Celine Grenier
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The Universtiy of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - David Severs
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| | - Alicja Czopek
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The Universtiy of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Jessica R Ivy
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The Universtiy of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Mark Nixon
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The Universtiy of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Megan C Holmes
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The Universtiy of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Dawn E W Livingstone
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The Universtiy of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| | - Neeraj Dhaun
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The Universtiy of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Matthew A Bailey
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The Universtiy of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| |
Collapse
|
9
|
Ahmad T, Ertuglu LA, Masenga SK, Kleyman TR, Kirabo A. The epithelial sodium channel in inflammation and blood pressure modulation. Front Cardiovasc Med 2023; 10:1130148. [PMID: 37123470 PMCID: PMC10132033 DOI: 10.3389/fcvm.2023.1130148] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
A major regulator of blood pressure and volume homeostasis in the kidney is the epithelial sodium channel (ENaC). ENaC is composed of alpha(α)/beta(β)/gamma(γ) or delta(δ)/beta(β)/gamma(γ) subunits. The δ subunit is functional in the guinea pig, but not in routinely used experimental rodent models including rat or mouse, and thus remains the least understood of the four subunits. While the δ subunit is poorly expressed in the human kidney, we recently found that its gene variants are associated with blood pressure and kidney function. The δ subunit is expressed in the human vasculature where it may influence vascular function. Moreover, we recently found that the δ subunit is also expressed human antigen presenting cells (APCs). Our studies indicate that extracellular Na+ enters APCs via ENaC leading to inflammation and salt-induced hypertension. In this review, we highlight recent findings on the role of extra-renal ENaC in inflammation, vascular dysfunction, and blood pressure modulation. Targeting extra-renal ENaC may provide new drug therapies for salt-induced hypertension.
Collapse
Affiliation(s)
- Taseer Ahmad
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Lale A. Ertuglu
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sepiso K. Masenga
- Department of Physiological Sciences, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
| | - Thomas R. Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
10
|
Power G, Padilla J. (Re)modeling high-salt diet-induced hypertension in mice. Am J Physiol Heart Circ Physiol 2023; 324:H470-H472. [PMID: 36827228 DOI: 10.1152/ajpheart.00093.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Gavin Power
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States.,NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States.,NextGen Precision Health, University of Missouri, Columbia, Missouri, United States.,Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
| |
Collapse
|
11
|
Little R, Murali SK, Poulsen SB, Grimm PR, Assmus A, Cheng L, Ivy JR, Hoorn EJ, Matchkov V, Welling PA, Fenton RA. Dissociation of sodium-chloride cotransporter expression and blood pressure during chronic high dietary potassium supplementation. JCI Insight 2023; 8:156437. [PMID: 36719746 PMCID: PMC10077486 DOI: 10.1172/jci.insight.156437] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Dietary potassium (K+) supplementation is associated with a lowering effect in blood pressure (BP), but not all studies agree. Here, we examined the effects of short- and long-term K+ supplementation on BP in mice, whether differences depend on the accompanying anion or the sodium (Na+) intake and molecular alterations in the kidney that may underlie BP changes. Relative to the control diet, BP was higher in mice fed a high NaCl (1.57% Na+) diet for 7 weeks or fed a K+-free diet for 2 weeks. BP was highest on a K+-free/high NaCl diet. Commensurate with increased abundance and phosphorylation of the thiazide sensitive sodium-chloride-cotransporter (NCC) on the K+-free/high NaCl diet, BP returned to normal with thiazides. Three weeks of a high K+ diet (5% K+) increased BP (predominantly during the night) independently of dietary Na+ or anion intake. Conversely, 4 days of KCl feeding reduced BP. Both feeding periods resulted in lower NCC levels but in increased levels of cleaved (active) α and γ subunits of the epithelial Na+ channel ENaC. The elevated BP after chronic K+ feeding was reduced by amiloride but not thiazide. Our results suggest that dietary K+ has an optimal threshold where it may be most effective for cardiovascular health.
Collapse
Affiliation(s)
- Robert Little
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Søren B Poulsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Paul R Grimm
- Departments of Medicine, Nephrology and Physiology, Johns Hopkins School of Medicine, Baltimore, USA
| | - Adrienne Assmus
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Lei Cheng
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jessica R Ivy
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Ewout J Hoorn
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Paul A Welling
- Departments of Medicine, Nephrology and Physiology, Johns Hopkins School of Medicine, Baltimore, USA
| | - Robert A Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
12
|
Xie Y, Qi H, Peng W, Li B, Wen F, Zhang F, Zhang L. Higher Potassium Intake and Lower Sodium Intake May Help in Reducing CVD Risk by Lowering Salt Sensitivity of Blood Pressure in the Han Chinese Population. Nutrients 2022; 14:nu14204436. [PMID: 36297119 PMCID: PMC9607620 DOI: 10.3390/nu14204436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 03/10/2023] Open
Abstract
Sodium (Na) reduction with a parallel supplemental potassium (K) intake can prevent cardiovascular diseases (CVDs). The relationship of the urinary Na/K ratio and salt sensitivity of blood pressure (SSBP) with CVDs is not clearly explained. We assumed that the SSBP mediates the relationship between the Na/K ratio and CVDs. In total, 2055 subjects who had 24 h urine collected and SSBP determined were included in this study. CVD risk was estimated using the China-PAR equation. MediationMultivariate logistic regression was used to explore the associations between the Na/K ratio or SSBP with CVD risk. Mediation analysis using a logistic regression model was performed. Both the urinary Na/K ratio and SSBP were related to the estimated CVD risk (p < 0.05). The mediation analysis found that SSBP mediated approximately 12% of the association between Na/K ratio and CVD risk. Our findings indicate that higher K intake and lower Na intake may help in preventing CVD risk by reducing SSBP risk in individuals with normotension or stage-one hypertension.
Collapse
Affiliation(s)
- Yunyi Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Han Qi
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Wenjuan Peng
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Bingxiao Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Fuyuan Wen
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Fengxu Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Ling Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
- Correspondence: ; Tel.: +86-10-83911777
| |
Collapse
|
13
|
Brown RB. Hypertension, Anxiety and Obstructive Sleep Apnea in Cardiovascular Disease and COVID-19: Mediation by Dietary Salt. Diseases 2022; 10:diseases10040089. [PMID: 36278588 PMCID: PMC9590013 DOI: 10.3390/diseases10040089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 12/02/2022] Open
Abstract
This perspective paper used a grounded theory method to synthesize evidence proposing that sodium toxicity from excessive dietary salt intake is a potential common pathophysiological mechanism that mediates the association of hypertension, obstructive sleep apnea, and anxiety with cardiovascular disease and COVID-19. Increased anxiety in these conditions may be linked to a high-salt diet through stimulation of the sympathetic nervous system, which increases blood pressure while releasing catecholamines, causing a "fight or flight" response. A rostral shift of fluid overload from the lower to the upper body occurs in obstructive sleep apnea associated with COVID-19 and cardiovascular disease, and may be related to sodium and fluid retention triggered by hypertonic dehydration. Chronic activation of the renin-angiotensin-aldosterone system responds to salt-induced dehydration by increasing reabsorption of sodium and fluid, potentially exacerbating fluid overload. Anxiety may also be related to angiotensin II that stimulates the sympathetic nervous system to release catecholamines. More research is needed to investigate these proposed interrelated mechanisms mediated by dietary salt. Furthermore, dietary interventions should use a whole-food plant-based diet that eliminates foods processed with salt to test the effect of very low sodium intake levels on hypertension, anxiety, and obstructive sleep apnea in cardiovascular disease and COVID-19.
Collapse
Affiliation(s)
- Ronald B Brown
- School of Public Health Sciences, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
14
|
Tissue Sodium Accumulation: Pathophysiology and Clinical Implications. Antioxidants (Basel) 2022; 11:antiox11040750. [PMID: 35453435 PMCID: PMC9031161 DOI: 10.3390/antiox11040750] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/02/2022] [Accepted: 04/07/2022] [Indexed: 02/05/2023] Open
Abstract
Excessive sodium intake has been well established as a risk factor for the development and progression of cardiovascular and renal diseases. Its adverse effects are achieved by renal sodium retention and related volume expansion and by inducing low-grade inflammation and oxidative stress (OS) in the target tissues. This review presents the recent concept of nonosmotic sodium storage in the skin interstitium, the subsequent dissociation of sodium and volume homeostasis, and the cellular response to the increased tissue sodium concentration. Furthermore, data are shown on the sodium barrier and buffering potential of the endothelial glycocalyx that may protect the functional integrity of the endothelium when it is challenged by an increased sodium load. Finally, examples will be given of the involvement of oxygen free radicals (OFR) in sodium-induced tissue damage, and some clinical entities will be mentioned that are causally associated with sodium/volume retention and OS.
Collapse
|
15
|
Rajanathan R, Pedersen TM, Thomsen MB, Botker HE, Matchkov VV. Phenylephrine-Induced Cardiovascular Changes in the Anesthetized Mouse: An Integrated Assessment of in vivo Hemodynamics Under Conditions of Controlled Heart Rate. Front Physiol 2022; 13:831724. [PMID: 35250634 PMCID: PMC8891648 DOI: 10.3389/fphys.2022.831724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Investigating the cardiovascular system is challenging due to its complex regulation by humoral and neuronal factors. Despite this complexity, many existing research methods are limited to the assessment of a few parameters leading to an incomplete characterization of cardiovascular function. Thus, we aim to establish a murine in vivo model for integrated assessment of the cardiovascular system under conditions of controlled heart rate. Utilizing this model, we assessed blood pressure, cardiac output, stroke volume, total peripheral resistance, and electrocardiogram (ECG). Hypothesis We hypothesize that (i) our in vivo model can be utilized to investigate cardiac and vascular responses to pharmacological intervention with the α1-agonist phenylephrine, and (ii) we can study cardiovascular function during artificial pacing of the heart, modulating cardiac function without a direct vascular effect. Methods We included 12 mice that were randomly assigned to either vehicle or phenylephrine intervention through intraperitoneal administration. Mice were anesthetized with isoflurane and intubated endotracheally for mechanical ventilation. We measured blood pressure via a solid-state catheter in the aortic arch, blood flow via a probe on the ascending aorta, and ECG from needle electrodes on the extremities. Right atrium was electrically paced at a frequency ranging from 10 to 11.3 Hz before and after either vehicle or phenylephrine administration. Results Phenylephrine significantly increased blood pressure, stroke volume, and total peripheral resistance compared to the vehicle group. Moreover, heart rate was significantly decreased following phenylephrine administration. Pacing significantly decreased stroke volume and cardiac output both prior to and after drug administration. However, phenylephrine-induced changes in blood pressure and total peripheral resistance were maintained with increasing pacing frequencies compared to the vehicle group. Total peripheral resistance was not significantly altered with increasing pacing frequencies suggesting that the effect of phenylephrine is primarily of vascular origin. Conclusion In conclusion, this in vivo murine model is capable of distinguishing between changes in peripheral vascular and cardiac functions. This study underlines the primary effect of phenylephrine on vascular function with secondary changes to cardiac function. Hence, this in vivo model is useful for the integrated assessment of the cardiovascular system.
Collapse
Affiliation(s)
- Rajkumar Rajanathan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- *Correspondence: Rajkumar Rajanathan,
| | | | - Morten B. Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Erik Botker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
16
|
Hunter RW, Dhaun N, Bailey MA. The impact of excessive salt intake on human health. Nat Rev Nephrol 2022; 18:321-335. [DOI: 10.1038/s41581-021-00533-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2021] [Indexed: 12/19/2022]
|
17
|
Culshaw G, Binnie D, Dhaun N, Hadoke P, Bailey M, Webb D. The acute pressure natriuresis response is suppressed by selective ETA receptor blockade. Clin Sci (Lond) 2021; 136:CS20210937. [PMID: 34918049 PMCID: PMC8734438 DOI: 10.1042/cs20210937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/25/2021] [Accepted: 12/16/2021] [Indexed: 11/17/2022]
Abstract
Hypertension is a major risk factor for cardiovascular disease. In a significant minority of people, it develops when salt intake is increased (salt-sensitivity). It is not clear whether this represents impaired vascular function or disruption to the relationship between blood pressure (BP) and renal salt-handling (pressure natriuresis, PN). Endothelin-1 (ET-1) regulates BP via ETA and ETB receptor subtypes. Blockade of ETA receptors reduces BP, but promotes sodium retention by an unknown mechanism. ETB blockade increases both BP and sodium retention. We hypothesised that ETA blockade promotes sodium and water retention by suppressing PN. We also investigated whether suppression of PN might reflect off-target ETB blockade. Acute PN was induced by sequential arterial ligation in male Sprague Dawley rats. Intravenous atrasentan (ETA antagonist, 5mg/kg) halved the normal increase in medullary perfusion and reduced sodium and water excretion by >60%. This was not due to off-target ETB blockade because intravenous A-192621 (ETB antagonist, 10mg/kg) increased natriuresis by 50% without modifying medullary perfusion. In a separate experiment in salt-loaded rats monitored by radiotelemetry, oral atrasentan reduced systolic and diastolic BP by ~10mmHg, but additional oral A-192621 reversed these effects. Endogenous ETA stimulation has natriuretic effects mediated by renal vascular dilation while endogenous ETB stimulation in the kidney has antinatriuretic effects via renal tubular mechanisms. Pharmacological manipulation of vascular function with ET antagonists modifies the BP set-point, but even highly selective ETA antagonists attenuate PN, which may be associated with salt and water retention.
Collapse
Affiliation(s)
- Geoffrey J. Culshaw
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - David Binnie
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - Neeraj Dhaun
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - Patrick W.F. Hadoke
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - Matthew A. Bailey
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - David J. Webb
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, U.K
| |
Collapse
|
18
|
Single-cell analysis of salt-induced hypertensive mouse aortae reveals cellular heterogeneity and state changes. Exp Mol Med 2021; 53:1866-1876. [PMID: 34862465 PMCID: PMC8741768 DOI: 10.1038/s12276-021-00704-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/26/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022] Open
Abstract
Elevated blood pressure caused by excessive salt intake is common and associated with cardiovascular diseases in most countries. However, the composition and responses of vascular cells in the progression of hypertension have not been systematically described. We performed single-cell RNA sequencing on the aortic arch from C57BL/6J mice fed a chow/high-salt diet. We identified 19 distinct cell populations representing 12 lineages, including smooth muscle cells (SMCs), fibroblasts, endothelial cells (ECs), B cells, and T cells. During the progression of hypertension, the proportion of three SMC subpopulations, two EC subpopulations, and T cells increased. In two EC clusters, the expression of reactive oxygen species-related enzymes, collagen and contractility genes was upregulated. Gene set enrichment analysis showed that three SMC subsets underwent endothelial-to-mesenchymal transition. We also constructed intercellular networks and found more frequent cell communication among aortic cells in hypertension and that some signaling pathways were activated during hypertension. Finally, joint public genome-wide association study data and our single-cell RNA-sequencing data showed the expression of hypertension susceptibility genes in ECs, SMCs, and fibroblasts and revealed 21 genes involved in the initiation and development of high-salt-induced hypertension. In conclusion, our data illustrate the transcriptional landscape of vascular cells in the aorta associated with hypertension and reveal dramatic changes in cell composition and intercellular communication during the progression of hypertension.
Collapse
|
19
|
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: 5.7] [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
|
20
|
Nakagawa P, Gomez J, Lu KT, Grobe JL, Sigmund CD. Studies of salt and stress sensitivity on arterial pressure in renin-b deficient mice. PLoS One 2021; 16:e0250807. [PMID: 34319999 PMCID: PMC8318244 DOI: 10.1371/journal.pone.0250807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/15/2021] [Indexed: 11/19/2022] Open
Abstract
Excessive sodium intake is known to increase the risk for hypertension, heart disease, and stroke. Individuals who are more susceptible to the effects of high salt are at higher risk for cardiovascular diseases even independent of their blood pressure status. Local activation of the renin-angiotensin system (RAS) in the brain, among other mechanisms, has been hypothesized to play a key role in contributing to salt balance. We have previously shown that deletion of the alternative renin isoform termed renin-b disinhibits the classical renin-a encoding preprorenin in the brain resulting in elevated brain RAS activity. Thus, we hypothesized that renin-b deficiency results in higher susceptibility to salt-induced elevation in blood pressure. Telemetry implanted Ren-bNull and wildtype littermate mice were first offered a low salt diet for a week and subsequently a high salt diet for another week. A high salt diet induced a mild blood pressure elevation in both Ren-bNull and wildtype mice, but mice lacking renin-b did not exhibit an exaggerated pressor response. When renin-b deficient mice were exposed to a high salt diet for a longer duration (4 weeks), there was a trend for increased myocardial enlargement in Ren-bNull mice when compared with control mice, but this did not reach statistical significance. Multiple studies have also demonstrated the association of environmental stress with hypertension. Activation of the RAS in the rostral ventrolateral medulla and the hypothalamus is required for stress-induced hypertension. Thus, we next questioned whether the lack of renin-b would result in exacerbated response to an acute restraint-stress. Wildtype and Ren-bNull mice equally exhibited elevated blood pressure in response to restraint-stress, which was similar in mice fed either a low or high salt diet. These studies suggest that mechanisms unrelated to salt and acute stress alter the cardiovascular phenotype in mice lacking renin-b.
Collapse
Affiliation(s)
- Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Javier Gomez
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Ko-Ting Lu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Curt D. Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| |
Collapse
|