1
|
Flores J, Pena C, Nugent K. Salt Sensitivity of Blood Pressure and the Role of the Immune System in Hypertension. Cardiol Rev 2024:00045415-990000000-00381. [PMID: 39679725 DOI: 10.1097/crd.0000000000000834] [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] [Indexed: 12/17/2024]
Abstract
Salt-sensitive blood pressure is a clinical phenotype defined as exaggerated blood pressure responses to salt loading and salt depletion. This characteristic occurs in 25% of the general population and 50% of patients with hypertension and contributes to the pathogenesis of hypertension in some patients. Hypertension is associated with chronic inflammatory responses and has immune cell accumulation in several hypertensive target organs, including the brain, kidneys, heart, blood vessels, and the perivascular adipose tissue, and these cellular responses likely exacerbate hypertension. The different factors implicated in the pathogenesis of salt-sensitive hypertension include renin-angiotensin-aldosterone system dysfunction, aldosterone-dependent and aldosterone-independent mineralocorticoid receptor signaling, and the sympathetic nervous system dysfunction. Experimental studies have shown an important role of both innate and adaptive immune cells, especially lymphocytes, in angiotensin II-induced hypertension. The epithelial sodium channel (ENaC) allows entry of sodium into dendritic cells, and this leads to a sequence of events, including the production of reactive oxygen species, which activates the NLRP3 inflammasome and contributes to salt-sensitive hypertension through the amiloride-sensitive ENaC and isolevuglandin-adduct formation. This review summarizes the general aspects of salt sensitivity, focuses on the immunological/inflammatory factors involved in its development, considers general changes in microvasculature, and discusses management.
Collapse
Affiliation(s)
- Jackeline Flores
- From the Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | | | | |
Collapse
|
2
|
Zahnow F, Jäger C, Mohamed Y, Vogelhuber G, May F, Ciocan AM, Manieri A, Maxeiner S, Krasteva-Christ G, Cobain MRD, Podsiadlowski L, Crespo-Picazo JL, García-Párraga D, Althaus M. The evolutionary path of the epithelial sodium channel δ-subunit in Cetartiodactyla points to a role in sodium sensing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.623996. [PMID: 39605611 PMCID: PMC11601428 DOI: 10.1101/2024.11.18.623996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The epithelial sodium channel (ENaC) plays a key role in osmoregulation in tetrapod vertebrates and is a candidate receptor for salt taste sensation. There are four ENaC subunits (α, β, γ, δ) which form αβγ- or δβγ ENaCs. While αβγ-ENaC is a 'maintenance protein' controlling sodium and potassium homeostasis, δβγ-ENaC might represent a 'stress protein' monitoring high sodium concentrations. The δ-subunit emerged with water-to-land transition of tetrapod vertebrate ancestors. We investigated the evolutionary path of ENaC-coding genes in Cetartiodactyla, a group comprising even-toed ungulates and the cetaceans (whales/dolphins) which transitioned from terrestrial to marine environments in the Eocene. The genes SCNN1A (α-ENaC), SCNN1B (β-ENaC) and SCNN1G (γ-ENaC) are intact in all 22 investigated cetartiodactylan families. While SCNN1D (δ-ENaC) is intact in terrestrial Artiodactyla, it is a pseudogene in 12 cetacean families. A fusion of SCNN1D exons 11 and 12 under preservation of the open reading frame was observed in the Antilopinae, representing a new feature of this clade. Transcripts of SCNN1A, SCNN1B and SCNN1G were present in kidney and lung tissues of Bottlenose dolphins, highlighting αβγ-ENaC's role as a maintenance protein. Consistent with SCNN1D loss, Bottlenose dolphins and Beluga whales did not show behavioural differences to stimuli with or without sodium in seawater-equivalent concentrations. These data suggest a function of δ-ENaC as a sodium sensing protein which might have become obsolete in cetaceans after the migration to high-salinity marine environments. Consistently, there is reduced selection pressure or pseudogenisation of SCNN1D in other marine mammals, including sirenians, pinnipeds and sea otter.
Collapse
Affiliation(s)
- Fynn Zahnow
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Chiara Jäger
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Yassmin Mohamed
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Gianluca Vogelhuber
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Fabian May
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Alexandra Maria Ciocan
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Arianna Manieri
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Stephan Maxeiner
- Institute of Anatomy and Cell Biology, Saarland University, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), Saarland University, Homburg, Germany
| | - Gabriela Krasteva-Christ
- Institute of Anatomy and Cell Biology, Saarland University, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), Saarland University, Homburg, Germany
| | - Matthew R. D. Cobain
- Department of Biological and Environmental Science, University of Jyväskylä, Finland
| | - Lars Podsiadlowski
- Leibniz Institute for the Analysis of Biodiversity Change (LIB), Bonn, Germany
| | | | - Daniel García-Párraga
- Research Department, Fundación Oceanogràfic de la Comunitat Valenciana, Valencia, Spain
| | - Mike Althaus
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| |
Collapse
|
3
|
Masenga SK, Desta S, Hatcher M, Kirabo A, Lee DL. How PPAR-alpha mediated inflammation may affect the pathophysiology of chronic kidney disease. Curr Res Physiol 2024; 8:100133. [PMID: 39665027 PMCID: PMC11629568 DOI: 10.1016/j.crphys.2024.100133] [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: 06/06/2024] [Revised: 10/03/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024] Open
Abstract
Chronic kidney disease (CKD) is a major risk factor for death in adults. Inflammation plays a role in the pathogenesis of CKD, but the mechanisms are poorly understood. Peroxisome proliferator-activated receptor alpha (PPAR-α) is a nuclear receptor and one of the three members (PPARα, PPARβ/δ, and PPARγ) of the PPARs that plays an important role in ameliorating pathological processes that accelerate acute and chronic kidney disease. Although other PPARs members are well studied, the role of PPAR-α is not well described and its role in inflammation-mediated chronic disease is not clear. Herein, we review the role of PPAR-α in chronic kidney disease with implications for the immune system.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Zambia
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Selam Desta
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| | - Mark Hatcher
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, 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
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dexter L. Lee
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| |
Collapse
|
4
|
Connolly BJ, Saxton SN. Recent updates on the influence of iron and magnesium on vascular, renal, and adipose inflammation and possible consequences for hypertension. J Hypertens 2024; 42:1848-1861. [PMID: 39258532 PMCID: PMC11451934 DOI: 10.1097/hjh.0000000000003829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/26/2024] [Accepted: 07/22/2024] [Indexed: 09/12/2024]
Abstract
The inflammatory status of the kidneys, vasculature, and perivascular adipose tissue (PVAT) has a significant influence on blood pressure and hypertension. Numerous micronutrients play an influential role in hypertension-driving inflammatory processes, and recent reports have provided bases for potential targeted modulation of these micronutrients to reduce hypertension. Iron overload in adipose tissue macrophages and adipocytes engenders an inflammatory environment and may contribute to impaired anticontractile signalling, and thus a treatment such as chelation therapy may hold a key to reducing blood pressure. Similarly, magnesium intake has proven to greatly influence inflammatory signalling and concurrent hypertension in both healthy animals and in a model for chronic kidney disease, demonstrating its potential clinical utility. These findings highlight the importance of further research to determine the efficacy of micronutrient-targeted treatments for the amelioration of hypertension and their potential translation into clinical application.
Collapse
Affiliation(s)
- Benjamin J Connolly
- Divison of Cardiovascular Sciences, The University of Manchester, Manchester, UK
| | | |
Collapse
|
5
|
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
|
6
|
Albritton CF, Hinton A, Kirabo A. Beyond the numbers in treating hypertensive end-organ damage: role of formyl peptide receptor agonist Cmpd17b. Cardiovasc Res 2024; 120:1239-1240. [PMID: 38980770 PMCID: PMC11416055 DOI: 10.1093/cvr/cvae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/11/2024] Open
Affiliation(s)
- Claude F Albritton
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 560 Preston Cancer Research Building, 2220 Pierce Ave, Nashville, TN 37232, USA
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
| | - Antentor Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 750 Robinson Research Building, 2200 Pierce Ave, Nashville, TN 37232-0615, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 560 Preston Cancer Research Building, 2220 Pierce Ave, Nashville, TN 37232, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 750 Robinson Research Building, 2200 Pierce Ave, Nashville, TN 37232-0615, USA
- Vanderbilt Center for Immunobiology, 1211 Medical Center Drive, Nashville, TN 37232-0615, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, 1161 21st Ave South Medical Center North A-5121, Nashville, TN 37232-0615, USA
- Vanderbilt Institute for Global Health, 2525 West End Avenue, Suite 750, Nashville, TN 37203, USA
| |
Collapse
|
7
|
Kirabo A. REPRIEVE and CANTOS trials: lessons learned towards eliminating cardiovascular disease in human immunodeficiency virus by combined targeting of LDL and inflammation. Cardiovasc Res 2024:cvae152. [PMID: 39250677 DOI: 10.1093/cvr/cvae152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/14/2024] [Accepted: 06/01/2024] [Indexed: 09/11/2024] Open
Affiliation(s)
- Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| |
Collapse
|
8
|
Xue J, Zhou Z, Zhu Z, Sun Q, Zhu Y, Wu P. A high salt diet impairs the bladder epithelial barrier and activates the NLRP3 and NF‑κB signaling pathways to induce an overactive bladder in vivo. Exp Ther Med 2024; 28:362. [PMID: 39071900 PMCID: PMC11273259 DOI: 10.3892/etm.2024.12651] [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: 01/23/2024] [Accepted: 05/22/2024] [Indexed: 07/30/2024] Open
Abstract
Overactive bladder (OAB) is a condition characterized by an urgency to urinate, which is associated with the urodynamic observation of detrusor overexcitation. Although the etiology of OAB is currently unclear, it has been suggested that in patients with OAB, disruption of bladder epithelial barrier integrity can disturb the normal contractile function of the detrusor. Additionally, dietary preferences have been suggested to influence the severity of OAB. Therefore, the aim of the present study was to investigate the effect of a high salt diet (HSD) on the development of OAB in a murine model. Mice were fed either a HSD or standard diet for 8 weeks, following which voiding characteristics and bladder barrier function were assessed. The present study demonstrated that a HSD in mice was associated with OAB-like symptoms such as increased urinary frequency and non-voiding bladder contractions. The HSD group demonstrated a thinner bladder mucus layer and decreased expression of bladder barrier markers, tight junction protein-1 and claudin-1, which may be potentially indicative of induced bladder damage. A HSD for 8 weeks in mice and a high salt treatment at the uroepithelium cellular (SV-HUC-1s) level resulted in increased uroepithelial oxidative stress and inflammatory cell infiltration, as indicated by increased expression levels of TNF-α and IL-1β, as well as activation of the nucleotide-binding domain leucine-rich-containing family pyrin domain-containing 3 (NLRP3) and NF-κB signaling pathways in vivo and in vitro. Therefore, the present study indicated that a HSD could be a potentially important risk factor for the development of OAB, as it may be associated with overactivation of contractile function of the bladder by impairing the integrity of the bladder epithelial barrier and activation of the NLRP3 and NF-κB signaling pathways. Remodeling of the bladder barrier and reduction of the inflammatory response may be potential targets for the treatment of OAB in the future.
Collapse
Affiliation(s)
- Jingwen Xue
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhipeng Zhou
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- Department of Urology, Jinshan Branch of Fujian Provincial Hospital, Fuzhou, Fujian 350004, P.R. China
| | - Zhangrui Zhu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qi Sun
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yuexuan Zhu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Peng Wu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
9
|
Chen Y, Li H, Zhang M, Wu Z, Fang H, Wen P, Zhang J, Guo W. Effects of donors' and recipients' preoperative serum sodium on the prognosis of liver transplantation. Sci Rep 2024; 14:20304. [PMID: 39218910 PMCID: PMC11366754 DOI: 10.1038/s41598-024-71218-4] [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: 03/12/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Dysnatremia is common in donors and recipients of liver transplantation (LT). However, the influence of dysnatremia on LT prognosis remains controversial. This study aimed to investigate effects of donors' and recipients' serum sodium on LT prognosis. We retrospectively reviewed 248 recipients who underwent orthotopic LT at our center between January 2016 and December 2018. Donors and recipients perioperative and 3-year postoperative clinical data were included. Delta serum sodium was defined as the donors' serum sodium minus the paired recipients' serum sodium. Donors with serum sodium > 145 mmol/L had significantly higher preoperative blood urea nitrogen (BUN) (P < 0.01) and creatinine (Cr) (P < 0.01) than others. Preoperative total bilirubin (TBIL) (P < 0.01), direct bilirubin (DBIL) (P < 0.01), BUN (P < 0.01), Cr (P < 0.01) were significantly higher in the hyponatremia group of recipients than the other groups, but both of donors' and recipients' serum sodium had no effect on the LT prognosis. In the delta serum sodium < 0 mmol/L group, TBIL (P < 0.01) and DBIL (P < 0.01) were significantly higher in postoperative 1 week than the other groups, but delta serum sodium had no effect on the postoperative survival rates. Dysnatremia in donors and recipients of LT have no effect on postoperative survival rates, hepatic and renal function, but recipients with higher serum sodium than donors have significantly higher TBIL and DBIL at 1 week postoperatively.
Collapse
Affiliation(s)
- Yabin Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, East Jian She Road, Zhengzhou, 450052, Henan, China
| | - Hao Li
- National Organ Transplantation (Liver &Kidney Transplantation) Physician Training Centre Zhengzhou, Henan, 450052, China
| | - Menggang Zhang
- National Regional Medical Treatment Centre of Henan Organ Transplantation, Zhengzhou, 450052, Henan, China
| | - Zeyu Wu
- Henan Organ Transplantation Centre, Zhengzhou, 450052, Henan, China
| | - Haoran Fang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, East Jian She Road, Zhengzhou, 450052, Henan, China
| | - Peihao Wen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, East Jian She Road, Zhengzhou, 450052, Henan, China
| | - Jiakai Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, East Jian She Road, Zhengzhou, 450052, Henan, China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, East Jian She Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
10
|
Shokoples BG, Paradis P, Schiffrin EL. Immunological insights into hypertension: unraveling triggers and potential therapeutic avenues. Hypertens Res 2024; 47:2115-2125. [PMID: 38778172 DOI: 10.1038/s41440-024-01731-6] [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: 01/27/2024] [Revised: 04/02/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Hypertension remains the leading cause of morbidity and mortality worldwide. Despite its prevalence, the development of novel antihypertensive therapies has only recently accelerated, with novel agents not yet commercialized, leaving a substantial proportion of individuals resistant to existing treatments. The intricate pathophysiology of hypertension is now understood to involve chronic low-grade inflammation, which places the immune system in the spotlight as a potential target for new therapeutics. This review explores the factors that initiate and sustain an immune response in hypertension, offering insights into potential targets for new treatments. Several factors contribute to immune activation in hypertension, including diet and damage-associated molecular pattern (DAMP) generation. Diets rich in fat or sodium can promote inflammation by inducing intestinal barrier dysfunction and triggering salt-sensitive receptors in T cells and dendritic cells. DAMPs, such as extracellular adenosine triphosphate and heat-shock protein 70, are released during episodes of increased blood pressure, contributing to immune cell activation and inflammation. Unconventional innate-like γδ T cells contribute to initiating and maintaining an immune response through their potential involvement in antigen presentation and regulating cytokine-mediated responses. Immunologic memory, sustained through the formation of effector memory T cells after exposure to hypertensive insults, likely contributes to maintaining an immune response in hypertension. When exposed to hypertensive insults, these memory cells are rapidly activated and contribute to elevated blood pressure and end-organ damage. Evidence from human hypertension, although limited, supports the relevance of distinct immune pathways in hypertension, and highlights the potential of targeted immune interventions in human hypertension. Diet and acute bouts of high blood pressure result in the release of dietary triggers, neoantigens, and damage-associated molecular patterns (DAMPs), which promote immune system activation. Elements such as lipopolysaccharides (LPS), sodium, heat-shock protein (HSP)70, extracellular adenosine triphosphate (eATP), and growth arrest-specific 6 (GAS6) promote activation of innate immune cells such as dendritic cells (DCs) and monocytes (Mo) through their respective receptors (toll-like receptor [TLR]4, amiloride-sensitive epithelial sodium channel [ENaC], TLR2/4, P2X7 receptor [P2RX7], and Axl) leading to costimulatory molecule expression and interleukin (IL)-1β and IL-23 production. The neoantigens HSP70 and isolevuglandins (IsoLGs) are presented to T cells by DCs and possibly γδ T cells, triggering T cell activation, IL-17 and interferon (IFN)-γ production, and the formation of T effector memory (TEM) cells in the kidney, perivascular adipose tissue, bone marrow, and spleen. Exposure of TEM cells to their cognate antigen or previous activating stimuli causes these cells rapid expansion and activation. Cumulatively, this inflammatory state contributes to hypertension and end-organ damage. The figure was created using images from smart.servier.com and is licensed under a Creative Commons Attribution 4.0 license (CC BY 4.0).
Collapse
Affiliation(s)
- Brandon G Shokoples
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research and McGill University, Montréal, QC, Canada
| | - Pierre Paradis
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research and McGill University, Montréal, QC, Canada
| | - Ernesto L Schiffrin
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research and McGill University, Montréal, QC, Canada.
- Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montréal, QC, Canada.
| |
Collapse
|
11
|
Zhang J, Liu S, Ding W, Wan J, Qin JJ, Wang M. Resolution of inflammation, an active process to restore the immune microenvironment balance: A novel drug target for treating arterial hypertension. Ageing Res Rev 2024; 99:102352. [PMID: 38857706 DOI: 10.1016/j.arr.2024.102352] [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: 11/24/2023] [Revised: 05/11/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024]
Abstract
The resolution of inflammation, the other side of the inflammatory response, is defined as an active and highly coordinated process that promotes the restoration of immune microenvironment balance and tissue repair. Inflammation resolution involves several key processes, including dampening proinflammatory signaling, specialized proresolving lipid mediator (SPM) production, nonlipid proresolving mediator production, efferocytosis and regulatory T-cell (Treg) induction. In recent years, increasing attention has been given to the effects of inflammation resolution on hypertension. Furthermore, our previous studies reported the antihypertensive effects of SPMs. Therefore, in this review, we aim to summarize and discuss the detailed association between arterial hypertension and inflammation resolution. Additional, the association between gut microbe-mediated immune and hypertension is discussed. This findings suggested that accelerating the resolution of inflammation can have beneficial effects on hypertension and its related organ damage. Exploring novel drug targets by focusing on various pathways involved in accelerating inflammation resolution will contribute to the treatment and control of hypertensive diseases in the future.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Siqi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China; Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
12
|
Nguyen BA, Alexander MR, Harrison DG. Immune mechanisms in the pathophysiology of hypertension. Nat Rev Nephrol 2024; 20:530-540. [PMID: 38658669 DOI: 10.1038/s41581-024-00838-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
Hypertension is a leading risk factor for morbidity and mortality worldwide. Despite current anti-hypertensive therapies, most individuals with hypertension fail to achieve adequate blood pressure control. Moreover, even with adequate control, a residual risk of cardiovascular events and associated organ damage remains. These findings suggest that current treatment modalities are not addressing a key element of the underlying pathology. Emerging evidence implicates immune cells as key mediators in the development and progression of hypertension. In this Review, we discuss our current understanding of the diverse roles of innate and adaptive immune cells in hypertension, highlighting key findings from human and rodent studies. We explore mechanisms by which these immune cells promote hypertensive pathophysiology, shedding light on their multifaceted involvement. In addition, we highlight advances in our understanding of autoimmunity, HIV and immune checkpoints that provide valuable insight into mechanisms of chronic and dysregulated inflammation in hypertension.
Collapse
Affiliation(s)
- Bianca A Nguyen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Matthew R Alexander
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, USA
| | - David G Harrison
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, USA.
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
13
|
Harrison DG, Patrick DM. Immune Mechanisms in Hypertension. Hypertension 2024; 81:1659-1674. [PMID: 38881474 PMCID: PMC11254551 DOI: 10.1161/hypertensionaha.124.21355] [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] [Indexed: 06/18/2024]
Abstract
It is now apparent that immune mediators including complement, cytokines, and cells of the innate and adaptive immune system contribute not only to blood pressure elevation but also to the target organ damage that occurs in response to stimuli like high salt, aldosterone, angiotensin II, and sympathetic outflow. Alterations of vascular hemodynamic factors, including microvascular pulsatility and shear forces, lead to vascular release of mediators that affect myeloid cells to become potent antigen-presenting cells and promote T-cell activation. Research in the past 2 decades has defined specific biochemical and molecular pathways that are engaged by these stimuli and an emerging paradigm is these not only lead to immune activation, but that products of immune cells, including cytokines, reactive oxygen species, and metalloproteinases act on target cells to further raise blood pressure in a feed-forward fashion. In this review, we will discuss these molecular and pathophysiological events and discuss clinical interventions that might prove effective in quelling this inflammatory process in hypertension and related cardiovascular diseases.
Collapse
Affiliation(s)
- David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - David M. Patrick
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Veterans Affairs, Nashville, TN 37212
| |
Collapse
|
14
|
Rahman S, Gamboa A, Saleem M, Kulapatana S, Diedrich A, Biaggioni I, Kirabo A, Shibao CA. Complete autonomic blockade reveals nitric oxide contribution to blood pressure regulation in obese Black women. Clin Auton Res 2024; 34:427-436. [PMID: 39090323 PMCID: PMC11362192 DOI: 10.1007/s10286-024-01050-3] [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: 01/25/2024] [Accepted: 06/26/2024] [Indexed: 08/04/2024]
Abstract
PURPOSE Hypertension is one of the major causes of cardiovascular morbidity and mortality in the USA and disproportionately affects Black women. Endothelial-derived nitric oxide (eNO) substantially regulates blood pressure in humans, and impaired NO-mediated vasodilation has been reported in the Black population. Previous studies using an NO synthase inhibitor, NG-monomethyl-L-arginine (L-NMMA) did not fully determine the NO contribution to blood pressure because of baroreflex buffering. Therefore, in the present study we used trimethaphan, a ganglionic blocker, to inhibit baroreflex buffering and study NO modulation of blood pressure in Black women during L-NMMA infusion. METHODS L-NMMA at doses of 250 μg/kg per minute was infused in combination with trimethaphan at doses of 4 mg/min to eliminate baroreflex mechanisms. Heart rate (HR) was obtained with continuous electrocardiogram monitoring, and continuous blood pressure was measured with the volume clamp method. The increase in systolic blood pressure (SBP) during both infusions was used to estimate the contribution of NO to blood pressure. RESULTS Ten Black (age range 30-50 years, body mass index [BMI] 30-45 kg/m2), and nine White women (age range 30-50 years, body mass index 30-45 kg/m2) were enrolled in this study. During autonomic blockade, there was no difference in the decrease in SBP between Black and White women (- 20 ± 16.45 vs. - 24 ± 15.49 mm Hg, respectively; P = 0.659). When autonomic blockade was combined with L-NMMA, Black women had a significant increase in SBP compared to White women (54 ± 13.62 vs. 39 ± 09.64 mm Hg, respectively; P = 0.022, respectively). CONCLUSION Autonomic blood pressure regulation was similar between Black and White women. However, NO contribution to blood pressure was significantly greater in Black women compared to White women. REGISTRATION ClinicalTrials.gov: NCT01122407.
Collapse
Affiliation(s)
- Sharla Rahman
- Department of Medicine, Epidemiology, Vanderbilt University School of Medicine, Nashville, TN, 37212-8802, USA
| | - Alfredo Gamboa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN, 37212-8802, USA
| | - Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN, 37212-8802, USA
| | - Surat Kulapatana
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN, 37212-8802, USA
- Department of Physiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - André Diedrich
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN, 37212-8802, USA
| | - Italo Biaggioni
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN, 37212-8802, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, P415C Medical Research Building IV, 2215 Garland Avenue, Nashville, TN, 37232, USA.
| | - Cyndya A Shibao
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN, 37212-8802, USA.
| |
Collapse
|
15
|
Dong W, Luo M, Li Y, Chen X, Li L, Chang Q. MICT ameliorates hypertensive nephropathy by inhibiting TLR4/NF-κB pathway and down-regulating NLRC4 inflammasome. PLoS One 2024; 19:e0306137. [PMID: 39052650 PMCID: PMC11271930 DOI: 10.1371/journal.pone.0306137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/11/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Hypertensive nephropathy (HN) is one of the main causes of end-stage renal disease (ESRD), leading to serious morbidity and mortality in hypertensive patients. However, existing treatment for hypertensive nephropathy are still very limited. It has been demonstrated that aerobic exercise has beneficial effects on the treatment of hypertension. However, the underlying mechanisms of exercise in HN remain unclear. METHODS The spontaneously hypertensive rats (SHR) were trained for 8 weeks on a treadmill with different exercise prescriptions. We detected the effects of moderate intensity continuous training (MICT) and high intensity interval training (HIIT) on inflammatory response, renal function, and renal fibrosis in SHR. We further investigated the relationship between TLR4 and the NLRC4 inflammasome in vitro HN model. RESULTS MICT improved renal fibrosis and renal injury, attenuating the inflammatory response by inhibiting TLR4/NF-κB pathway and the activation of NLRC4 inflammasome. However, these changes were not observed in the HIIT group. Additionally, repression of TLR4/NF-κB pathway by TAK-242 inhibited activation of NLRC4 inflammasome and alleviated the fibrosis in Ang II-induced HK-2 cells. CONCLUSION MICT ameliorated renal damage, inflammatory response, and renal fibrosis via repressing TLR4/NF-κB pathway and the activation of NLRC4 inflammasome. This study might provide new references for exercise prescriptions of hypertension.
Collapse
Affiliation(s)
- Wenyu Dong
- The Affiliated Rehabilitation Hospital, Chongqing Medical University, Chongqing, P. R. China
| | - Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Yun Li
- The Affiliated Rehabilitation Hospital, Chongqing Medical University, Chongqing, P. R. China
| | - Xinhua Chen
- The Affiliated Rehabilitation Hospital, Chongqing Medical University, Chongqing, P. R. China
| | - Lingang Li
- The Affiliated Rehabilitation Hospital, Chongqing Medical University, Chongqing, P. R. China
| | - Qing Chang
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
16
|
Masenga SK, Liweleya S, Kirabo A. High salt intake and HIV infection on endothelial glycocalyx shedding in salt-sensitive hypertension. Front Cell Dev Biol 2024; 12:1395885. [PMID: 39081863 PMCID: PMC11286502 DOI: 10.3389/fcell.2024.1395885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
The endothelial glycocalyx is closely associated with various physiological and pathophysiological events. Significant modification of the endothelial glycocalyx is an early process in the pathogenesis of cardiovascular disease. High dietary salt and HIV infection damages the endothelial glycocalyx causing endothelial dysfunction and increasing the risk for salt-sensitive hypertension and cardiovascular disease. The two factors, HIV infection and dietary salt are critical independent predictors of hypertension and cardiovascular disease and often synergize to exacerbate and accelerate disease pathogenesis. Salt-sensitive hypertension is more common among people living with HIV and is associated with risk for cardiovascular disease, stroke, heart attack and even death. However, the underlying mechanisms linking endothelial glycocalyx damage to dietary salt and HIV infection are lacking. Yet, both HIV infection/treatment and dietary salt are closely linked to endothelial glycocalyx damage and development of salt-sensitive hypertension. Moreover, the majority of individuals globally, consume more salt than is recommended and the burden of HIV especially in sub-Sahara Africa is disproportionately high. In this review, we have discussed the missing link between high salt and endothelial glycocalyx shedding in the pathogenesis of salt-sensitive hypertension. We have further elaborated the role played by HIV infection and treatment in modifying endothelial glycocalyx integrity to contribute to the development of hypertension and cardiovascular disease.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Situmbeko Liweleya
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, United States
- Vanderbilt Institute for Global Health, Nashville, TN, United States
| |
Collapse
|
17
|
Ertuglu LA, Sahinoz M, Alsouqi A, Deger SM, Guide A, Pike M, Robinson‐Cohen C, Akwo E, Pridmore M, Crescenzi R, Madhur MS, Kirabo A, Harrison DG, Luft FC, Titze J, Ikizler TA, Gamboa JL. Intermuscular adipose tissue accumulation is associated with higher tissue sodium in healthy individuals. Physiol Rep 2024; 12:e16127. [PMID: 38960895 PMCID: PMC11222016 DOI: 10.14814/phy2.16127] [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: 03/27/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND AND AIMS High tissue sodium accumulation and intermuscular adipose tissue (IMAT) are associated with aging, type 2 diabetes, and chronic kidney disease. In this study, we aim to investigate whether high lower-extremity tissue sodium accumulation relates to IMAT quantity and whether systemic inflammatory mediators and adipocytokines contribute to such association. METHODS Tissue sodium content and IMAT accumulation (percentage of IMAT area to muscle area) were measured in 83 healthy individuals using sodium imaging (23Na-MRI) and proton (1H-MRI) imaging of the calf. Insulin sensitivity was assessed by glucose disposal rate (GDR) measured with the hyperinsulinemic-euglycemic clamp. RESULTS Median (interquartile range) muscle and skin sodium contents were 16.6 (14.9, 19.0) and 12.6 (10.9, 16.7) mmol/L, respectively. Median IMAT was 3.69 (2.80, 5.37) %. In models adjusted for age, sex, BMI, GDR, adiponectin, and high-sensitivity C-reactive protein, increasing tissue sodium content was significantly associated with higher IMAT quantity (p = 0.018 and 0.032 for muscle and skin tissue sodium, respectively). In subgroup analysis stratified by sex, skin sodium was significantly associated with IMAT only among men. In interaction analysis, the association between skin sodium and IMAT was greater with increasing levels of high-sensitivity C-reactive protein and interleukin-6 (p for interaction = 0.022 and 0.006, respectively). CONCLUSIONS Leg muscle and skin sodium are associated with IMAT quantity among healthy individuals. The relationship between skin sodium and IMAT may be mediated by systemic inflammation.
Collapse
Affiliation(s)
- Lale A. Ertuglu
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Melis Sahinoz
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Aseel Alsouqi
- Now with Division of Hematology and Oncology, Department of MedicineUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Serpil Muge Deger
- Division of Nephrology, Department of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Andrew Guide
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Mindy Pike
- Division of Epidemiology, Department of MedicineVanderbilt UniversityNashvilleTennesseeUSA
| | - Cassianne Robinson‐Cohen
- Division of Nephrology and Hypertension, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Elvis Akwo
- Division of Nephrology and Hypertension, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Michael Pridmore
- Department of Radiology and Radiological SciencesVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Rachelle Crescenzi
- Department of Radiology and Radiological SciencesVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| | - Meena S. Madhur
- Division of Clinical Pharmacology, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Molecular Physiology and BiophysicsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - David G. Harrison
- Division of Clinical Pharmacology, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | | | - Jens Titze
- Program in Cardiovascular and Metabolic DisordersDuke NUS Medical SchoolBukit MerahSingapore
| | - T. Alp Ikizler
- Division of Nephrology and Hypertension, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jorge L. Gamboa
- Division of Clinical Pharmacology, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
18
|
Albritton CF, Demirci M, Neikirk K, Ertuglu LA, Ishimwe JA, Mutchler AL, Sheng Q, Laffer CL, Wanjalla CN, Ahmed T, Haynes AP, Saleem M, Beasley HK, Marshall AG, Vue Z, Ikizler AT, Kleyman TR, Kon V, Hinton A, Kirabo A. Myeloid Cell Glucocorticoid, Not Mineralocorticoid Receptor Signaling, Contributes to Salt-Sensitive Hypertension in Humans via Cortisol. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598374. [PMID: 38915603 PMCID: PMC11195113 DOI: 10.1101/2024.06.10.598374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
BACKGROUND Salt sensitivity of blood pressure (SSBP) is an independent risk factor for cardiovascular morbidity and mortality, yet the etiology is poorly understood. We previously found that serum/glucocorticoid-regulated kinase 1 (SGK1) and epoxyeicosatrienoic acids (EETs) regulate epithelial sodium channel (ENaC)-dependent sodium entry into monocyte-derived antigen-presenting cells (APCs) and activation of NADPH oxidase, leading to the formation of isolevuglandins (IsoLGs) in SSBP. Whereas aldosterone via the mineralocorticoid receptor (MR) activates SGK1 leading to hypertension, our past findings indicate that levels of plasma aldosterone do not correlate with SSBP, and there is little to no MR expression in APCs. Thus, we hypothesized that cortisol acting via the glucocorticoid receptor (GR), not the MR in APCs mediates SGK1 actions to induce SSBP. METHODS We performed cellular indexing of transcriptomes and epitopes by sequencing (CITE-Seq) analysis on peripheral blood mononuclear cells of humans rigorously phenotyped for SSBP using an inpatient salt loading/depletion protocol to determine expression of MR, GR, and SGK1 in immune cells. In additional experiments, we performed bulk transcriptomic analysis on isolated human monocytes following in vitro treatment with high salt from a separate cohort. We then measured urine and plasma cortisol, cortisone, renin, and aldosterone. Subsequently, we measured the association of these hormones with changes in systolic, diastolic, mean arterial pressure and pulse pressure as well as immune cell activation via IsoLG formation. RESULTS We found that myeloid APCs predominantly express the GR and SGK1 with no expression of the MR. Expression of the GR in APCs increased after salt loading and decreased with salt depletion in salt-sensitive but not salt-resistant people and was associated with increased expression of SGK1. Moreover, we found that plasma and urine cortisol/cortisone but not aldosterone/renin correlated with SSBP and APCs activation via IsoLGs. We also found that cortisol negatively correlates with EETs. CONCLUSION Our findings suggest that renal cortisol signaling via the GR but not the MR in APCs contributes to SSBP via cortisol. Urine and plasma cortisol may provide an important currently unavailable feasible diagnostic tool for SSBP. Moreover, cortisol-GR-SGK1-ENaC signaling pathway may provide treatment options for SSBP.
Collapse
Affiliation(s)
- Claude F. Albritton
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
| | - Mert Demirci
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kit Neikirk
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Lale A. Ertuglu
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jeanne A Ishimwe
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Ashley L Mutchler
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Cheryl L Laffer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Celestine N. Wanjalla
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Taseer Ahmed
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Pharmacology, College of Pharmacy, University of Sargodha, University Road, Sargodha, Punjab, 40100, Pakistan
| | - Alexandria Porcia Haynes
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Heather K. Beasley
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Andrea G. Marshall
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Zer Vue
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Alp T Ikizler
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Thomas R. Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Valentina Kon
- Division of Nephrology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Antentor Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health
| |
Collapse
|
19
|
Balhara M, Neikirk K, Marshall A, Hinton A, Kirabo A. Endoplasmic Reticulum Stress in Hypertension and Salt Sensitivity of Blood Pressure. Curr Hypertens Rep 2024; 26:273-290. [PMID: 38602583 PMCID: PMC11166838 DOI: 10.1007/s11906-024-01300-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2024] [Indexed: 04/12/2024]
Abstract
PURPOSE OF REVIEW Hypertension is a principal risk factor for cardiovascular morbidity and mortality, with its severity exacerbated by high sodium intake, particularly in individuals with salt-sensitive blood pressure. However, the mechanisms underlying hypertension and salt sensitivity are only partly understood. Herein, we review potential interactions in hypertension pathophysiology involving the immune system, endoplasmic reticulum (ER) stress, the unfolded protein response (UPR), and proteostasis pathways; identify knowledge gaps; and discuss future directions. RECENT FINDINGS Recent advancements by our research group and others reveal interactions within and between adaptive and innate immune responses in hypertension pathophysiology. The salt-immune-hypertension axis is further supported by the discovery of the role of dendritic cells in hypertension, marked by isolevuglandin (IsoLG) formation. Alongside these broadened understandings of immune-mediated salt sensitivity, the contributions of T cells to hypertension have been recently challenged by groups whose findings did not support increased resistance of Rag-1-deficient mice to Ang II infusion. Hypertension has also been linked to ER stress and the UPR. Notably, a holistic approach is needed because the UPR engages in crosstalk with autophagy, the ubiquitin proteasome, and other proteostasis pathways, that may all involve hypertension. There is a critical need for studies to establish cause and effect relationships between ER stress and the UPR in hypertension pathophysiology in humans and to determine whether the immune system and ER stress function mainly to exacerbate or initiate hypertension and target organ injury. This review of recent studies proposes new avenues for future research for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Maria Balhara
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Andrea Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA.
- Vanderbilt Center for Immunobiology, Nashville, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, USA.
- Vanderbilt Institute for Global Health, Nashville, USA.
| |
Collapse
|
20
|
Guzik TJ, Nosalski R, Maffia P, Drummond GR. Immune and inflammatory mechanisms in hypertension. Nat Rev Cardiol 2024; 21:396-416. [PMID: 38172242 DOI: 10.1038/s41569-023-00964-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 01/05/2024]
Abstract
Hypertension is a global health problem, with >1.3 billion individuals with high blood pressure worldwide. In this Review, we present an inflammatory paradigm for hypertension, emphasizing the crucial roles of immune cells, cytokines and chemokines in disease initiation and progression. T cells, monocytes, macrophages, dendritic cells, B cells and natural killer cells are all implicated in hypertension. Neoantigens, the NLRP3 inflammasome and increased sympathetic outflow, as well as cytokines (including IL-6, IL-7, IL-15, IL-18 and IL-21) and a high-salt environment, can contribute to immune activation in hypertension. The activated immune cells migrate to target organs such as arteries (especially the perivascular fat and adventitia), kidneys, the heart and the brain, where they release effector cytokines that elevate blood pressure and cause vascular remodelling, renal damage, cardiac hypertrophy, cognitive impairment and dementia. IL-17 secreted by CD4+ T helper 17 cells and γδ T cells, and interferon-γ and tumour necrosis factor secreted by immunosenescent CD8+ T cells, exert crucial effector roles in hypertension, whereas IL-10 and regulatory T cells are protective. Effector mediators impair nitric oxide bioavailability, leading to endothelial dysfunction and increased vascular contractility. Inflammatory effector mediators also alter renal sodium and water balance and promote renal fibrosis. These mechanisms link hypertension with obesity, autoimmunity, periodontitis and COVID-19. A comprehensive understanding of the immune and inflammatory mechanisms of hypertension is crucial for safely and effectively translating the findings to clinical practice.
Collapse
Affiliation(s)
- Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK.
- Department of Medicine and Omicron Medical Genomics Laboratory, Jagiellonian University, Collegium Medicum, Kraków, Poland.
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK.
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Pasquale Maffia
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Grant R Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Krishnan J, Hennen EM, Ao M, Kirabo A, Ahmad T, de la Visitación N, Patrick DM. NETosis Drives Blood Pressure Elevation and Vascular Dysfunction in Hypertension. Circ Res 2024; 134:1483-1494. [PMID: 38666386 PMCID: PMC11116040 DOI: 10.1161/circresaha.123.323897] [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: 10/25/2023] [Revised: 03/18/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Neutrophil extracellular traps (NETs) are composed of DNA, enzymes, and citrullinated histones that are expelled by neutrophils in the process of NETosis. NETs accumulate in the aorta and kidneys in hypertension. PAD4 (protein-arginine deiminase-4) is a calcium-dependent enzyme that is essential for NETosis. TRPV4 (transient receptor potential cation channel subfamily V member 4) is a mechanosensitive calcium channel expressed in neutrophils. Thus, we hypothesize that NETosis contributes to hypertension via NET-mediated endothelial cell (EC) dysfunction. METHODS NETosis-deficient Padi4-/- mice were treated with Ang II (angiotensin II). Blood pressure was measured by radiotelemetry, and vascular reactivity was measured with wire myography. Neutrophils were cultured with or without ECs and exposed to normotensive or hypertensive uniaxial stretch. NETosis was measured by flow cytometry. ECs were treated with citrullinated histone H3, and gene expression was measured by quantitative reverse transcription PCR. Aortic rings were incubated with citrullinated histone H3, and wire myography was performed to evaluate EC function. Neutrophils were treated with the TRPV4 agonist GSK1016790A. Calcium influx was measured using Fluo-4 dye, and NETosis was measured by immunofluorescence. RESULTS Padi4-/- mice exhibited attenuated hypertension, reduced aortic inflammation, and improved EC-dependent vascular relaxation in response to Ang II. Coculture of neutrophils with ECs and exposure to hypertensive uniaxial stretch increased NETosis and accumulation of neutrophil citrullinated histone H3. Histone H3 and citrullinated histone H3 exposure attenuates EC-dependent vascular relaxation. Treatment of neutrophils with the TRPV4 agonist GSK1016790A increases intracellular calcium and NETosis. CONCLUSIONS These observations identify a role of NETosis in the pathogenesis of hypertension. Moreover, they define an important role of EC stretch and TRPV4 as initiators of NETosis. Finally, they define a role of citrullinated histones as drivers of EC dysfunction in hypertension.
Collapse
Affiliation(s)
- Jaya Krishnan
- Division of Clinical Pharmacology, Department of Medicine (J.K., A.K., T.A., N.d.l.V., D.M.P.), Vanderbilt University Medical Center, Nashville, TN
| | - Elizabeth M. Hennen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN (E.M.H.)
| | - Mingfang Ao
- Department of Anesthesiology (M.A.), Vanderbilt University Medical Center, Nashville, TN
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine (J.K., A.K., T.A., N.d.l.V., D.M.P.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN (A.K.)
- Vanderbilt Institute for Global Health, Nashville, TN (A.K.)
| | - Taseer Ahmad
- Division of Clinical Pharmacology, Department of Medicine (J.K., A.K., T.A., N.d.l.V., D.M.P.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan (T.A.)
| | - Néstor de la Visitación
- Division of Clinical Pharmacology, Department of Medicine (J.K., A.K., T.A., N.d.l.V., D.M.P.), Vanderbilt University Medical Center, Nashville, TN
- Division of Cardiovascular Medicine, Department of Medicine (D.M.P.), Vanderbilt University Medical Center, Nashville, TN
| | - David M. Patrick
- Division of Clinical Pharmacology, Department of Medicine (J.K., A.K., T.A., N.d.l.V., D.M.P.), Vanderbilt University Medical Center, Nashville, TN
- Department of Veterans Affairs, Nashville, TN (D.M.P.)
| |
Collapse
|
22
|
Abstract
This interdisciplinary review explores the intricate nexus between HIV infection, nutrition, adrenal gland function, and cardiovascular health, highlighting a critical aspect of HIV management often overlooked in current literature. With the advent of antiretroviral therapy, the life expectancy of people living with HIV has dramatically improved, transforming HIV into a manageable chronic condition. However, this success brings forth new challenges, notably an increased risk of cardiovascular diseases among people living with HIV. We examine the normal physiology of the adrenal gland, including its role in mineral metabolism, a crucial facet of nutrition. We discuss the evolution of knowledge tying adrenal pathology to cardiovascular disease. We explore the impact of HIV on adrenal gland findings from a gross pathology perspective, as well as the clinical impact of adrenal insufficiency in HIV. The review further elucidates the role of nutrition in this context, considering the double burden of undernutrition and obesity prevalent in regions heavily affected by HIV. By aggregating findings from longitudinal studies and recent clinical trials, the review presents compelling evidence of increased cardiovascular disease among people living with HIV compared with people without HIV. It highlights the critical role of the adrenal glands in regulating nutrient metabolism and its implications for cardiovascular health, drawing attention to the potential for dietary interventions and targeted therapies to mitigate these risks. This review urges a paradigm shift in the management of HIV, advocating for a holistic approach that incorporates nutritional assessment and interventions into routine HIV care to address the complex interplay between HIV, adrenal function, and cardiovascular health. Through this lens, we offer insights into novel therapeutic strategies aimed at reducing cardiovascular risk in people living with HIV, contributing to the ongoing efforts to enhance the quality of life and longevity in this population.
Collapse
Affiliation(s)
- Anxious J Niwaha
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine, Uganda Research Unit, Entebbe (A.J.N.)
| | - James Brian Byrd
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (J.B.B.)
| |
Collapse
|
23
|
Khan MM, Kirabo A. Long Noncoding RNA MALAT1: Salt-Sensitive Hypertension. Int J Mol Sci 2024; 25:5507. [PMID: 38791545 PMCID: PMC11122212 DOI: 10.3390/ijms25105507] [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: 03/14/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Hypertension stands as the leading global cause of mortality, affecting one billion individuals and serving as a crucial risk indicator for cardiovascular morbidity and mortality. Elevated salt intake triggers inflammation and hypertension by activating antigen-presenting cells (APCs). We found that one of the primary reasons behind this pro-inflammatory response is the epithelial sodium channel (ENaC), responsible for transporting sodium ions into APCs and the activation of NADPH oxidase, leading to increased oxidative stress. Oxidative stress increases lipid peroxidation and the formation of pro-inflammatory isolevuglandins (IsoLG). Long noncoding RNAs (lncRNAs) play a crucial role in regulating gene expression, and MALAT1, broadly expressed across cell types, including blood vessels and inflammatory cells, is also associated with inflammation regulation. In hypertension, the decreased transcriptional activity of nuclear factor erythroid 2-related factor 2 (Nrf2 or Nfe2l2) correlates with heightened oxidative stress in APCs and impaired control of various antioxidant genes. Kelch-like ECH-associated protein 1 (Keap1), an intracellular inhibitor of Nrf2, exhibits elevated levels of hypertension. Sodium, through an increase in Sp1 transcription factor binding at its promoter, upregulates MALAT1 expression. Silencing MALAT1 inhibits sodium-induced Keap1 upregulation, facilitating the nuclear translocation of Nrf2 and subsequent antioxidant gene transcription. Thus, MALAT1, acting via the Keap1-Nrf2 pathway, modulates antioxidant defense in hypertension. This review explores the potential role of the lncRNA MALAT1 in controlling the Keap1-Nrf2-antioxidant defense pathway in salt-induced hypertension. The inhibition of MALAT1 holds therapeutic potential for the progression of salt-induced hypertension and cardiovascular disease (CVD).
Collapse
Affiliation(s)
- Mohd Mabood Khan
- Department of Medicine, Preston Research Building, Vanderbilt University Medical Centre, Nashville, TN 37232, USA
| | - Annet Kirabo
- Department of Medicine, Preston Research Building, Vanderbilt University Medical Centre, Nashville, TN 37232, USA
| |
Collapse
|
24
|
Afolabi J, Laffer CL, Beasley HK, Hinton A, Masenga SK, Kirabo A. Salt Sensitivity of Blood Pressure. Circ Res 2024; 134:1234-1239. [PMID: 38723029 DOI: 10.1161/circresaha.123.322982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/15/2024]
Abstract
The year 2024 marks the centennial of the initiation of the American Heart Association. Over the past 100 years, the American Heart Association has led groundbreaking discoveries in cardiovascular disease including salt sensitivity of blood pressure, which has been studied since the mid-1900s. Salt sensitivity of blood pressure is an important risk factor for cardiovascular events, but the phenotype remains unclear because of insufficient understanding of the underlying mechanisms and lack of feasible diagnostic tools. In honor of this centennial, we commemorate the initial discovery of salt sensitivity of blood pressure and chronicle the subsequent scientific discoveries and efforts to mitigate salt-induced cardiovascular disease with American Heart Association leading the way. We also highlight determinants of the pathophysiology of salt sensitivity of blood pressure in humans and recent developments in diagnostic methods and future prospects.
Collapse
Affiliation(s)
- Jeremiah Afolabi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.A., C.L.L., A.K.)
| | - Cheryl L Laffer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.A., C.L.L., A.K.)
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (H.K.B., A.H.)
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (H.K.B., A.H.)
| | - Sepiso K Masenga
- HAND Research Group (Hypertension, HIV/AIDS, Nutrition, Diabetes and Dyslipidemia Research Group), School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia (S.K.M.)
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.A., C.L.L., A.K.)
- Vanderbilt Center for Immunobiology, Nashville, TN (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN (A.K.)
- Vanderbilt Institute for Global Health, Nashville, TN (A.K.)
| |
Collapse
|
25
|
Fularski P, Czarnik W, Frankenstein H, Gąsior M, Młynarska E, Rysz J, Franczyk B. Unveiling Selected Influences on Chronic Kidney Disease Development and Progression. Cells 2024; 13:751. [PMID: 38727287 PMCID: PMC11083010 DOI: 10.3390/cells13090751] [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: 03/13/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 05/13/2024] Open
Abstract
Currently, more and more people are suffering from chronic kidney disease (CKD). It is estimated that CKD affects over 10% of the population worldwide. This is a significant issue, as the kidneys largely contribute to maintaining homeostasis by, among other things, regulating blood pressure, the pH of blood, and the water-electrolyte balance and by eliminating unnecessary metabolic waste products from blood. What is more, this disease does not show any specific symptoms at the beginning. The development of CKD is predisposed by certain conditions, such as diabetes mellitus or hypertension. However, these disorders are not the only factors promoting the onset and progression of CKD. The primary purpose of this review is to examine renin-angiotensin-aldosterone system (RAAS) activity, transforming growth factor-β1 (TGF-β1), vascular calcification (VC), uremic toxins, and hypertension in the context of their impact on the occurrence and the course of CKD. We firmly believe that a deeper comprehension of the cellular and molecular mechanisms underlying CKD can lead to an enhanced understanding of the disease. In the future, this may result in the development of medications targeting specific mechanisms involved in the decline of kidney function. Our paper unveils the selected processes responsible for the deterioration of renal filtration abilities.
Collapse
Affiliation(s)
- Piotr Fularski
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.G.)
| | - Witold Czarnik
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.G.)
| | - Hanna Frankenstein
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.G.)
| | - Magdalena Gąsior
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.G.)
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.G.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.G.)
| |
Collapse
|
26
|
AlShanableh Z, Ray EC. Magnesium in hypertension: mechanisms and clinical implications. Front Physiol 2024; 15:1363975. [PMID: 38665599 PMCID: PMC11044701 DOI: 10.3389/fphys.2024.1363975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Hypertension is associated with increased risk of cardiovascular disease and death. Evidence suggests that Mg2+ depletion contributes to hypertension. It is estimated that 25% or more of the United States population experiences chronic, latent Mg2+ depletion. This review explores mechanisms by which Mg2+ influences blood pressure, modifying risk of hypertension and complicating its treatment. Mechanisms addressed include effects upon i) sympathetic tone, via the modulation of N-methyl-D-aspartate (NMDA) receptor and N-type Ca2+ channel activity, influencing catecholamine release from sympathetic nerve endings; ii) vascular tone, via alteration of L-type Ca2+ and endothelial nitric oxide synthase (eNOS) activity and prostacyclin release; iii) renal K+ handling, influencing systemic K+ balance and potentially indirectly influencing blood pressure; iv) aldosterone secretion from the adrenal cortex; and v) modulation of pro-hypertensive inflammatory processes in dendritic cells and macrophages, including activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome and stimulation of isolevuglandin (IsoLG) production. Discovery of these mechanisms has furthered our understanding of the pathogenesis of hypertension, with implications for treatment and has highlighted the role of Mg2+ balance in hypertension and cardiovascular disease.
Collapse
Affiliation(s)
| | - Evan C. Ray
- Renal-Electrolyte Division, UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
27
|
Ertuglu LA, Mutchler AP, Jamison S, Laffer CL, Saleem M, Blackwell DJ, Kryshtal DO, Sahinoz M, Sheng Q, Wanjalla CN, Pakala S, Justin Y, Gutierrez OM, Kleyman TR, Knollmann BC, Ikizler TA, Kirabo A. Eicosanoid-Regulated Myeloid ENaC and Isolevuglandin Formation in Human Salt-Sensitive Hypertension. Hypertension 2024; 81:516-529. [PMID: 37675576 PMCID: PMC10918035 DOI: 10.1161/hypertensionaha.123.21285] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/15/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND The mechanisms by which salt increases blood pressure in people with salt sensitivity remain unclear. Our previous studies found that high sodium enters antigen-presenting cells (APCs) via the epithelial sodium channel and leads to the production of isolevuglandins and hypertension. In the current mechanistic clinical study, we hypothesized that epithelial sodium channel-dependent isolevuglandin-adduct formation in APCs is regulated by epoxyeicosatrienoic acids (EETs) and leads to salt-sensitive hypertension in humans. METHODS Salt sensitivity was assessed in 19 hypertensive subjects using an inpatient salt loading and depletion protocol. Isolevuglandin-adduct accumulation in APCs was analyzed using flow cytometry. Gene expression in APCs was analyzed using cellular indexing of transcriptomes and epitopes by sequencing analysis of blood mononuclear cells. Plasma and urine EETs were measured using liquid chromatography-mass spectrometry. RESULTS Baseline isolevuglandin+ APCs correlated with higher salt-sensitivity index. Isolevuglandin+ APCs significantly decreased from salt loading to depletion with an increasing salt-sensitivity index. We observed that human APCs express the epithelial sodium channel δ subunit, SGK1 (salt-sensing kinase serum/glucocorticoid kinase 1), and cytochrome P450 2S1. We found a direct correlation between baseline urinary 14,15 EET and salt-sensitivity index, whereas changes in urinary 14,15 EET negatively correlated with isolevuglandin+ monocytes from salt loading to depletion. Coincubation with 14,15 EET inhibited high-salt-induced increase in isolevuglandin+ APC. CONCLUSIONS Isolevuglandin formation in APCs responds to acute changes in salt intake in salt-sensitive but not salt-resistant people with hypertension, and this may be regulated by renal 14,15 EET. Baseline levels of isolevuglandin+ APCs or urinary 14,15 EET may provide diagnostic tools for salt sensitivity without a protocol of salt loading.
Collapse
Affiliation(s)
- Lale A. Ertuglu
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ashley Pitzer Mutchler
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - S Jamison
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
- Meharry Medical College Nashville, Nashville, TN, United States
| | - Cheryl L. Laffer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Daniel J. Blackwell
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Dmytro O. Kryshtal
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Melis Sahinoz
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Department of Internal Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center Nashville, TN, USA
| | - Suman Pakala
- Department of Internal Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center Nashville, TN, USA
| | - Yu Justin
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Orlando M Gutierrez
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Thomas R. Kleyman
- Departments of Medicine, Cell Biology, Pharmacology, and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Björn C. Knollmann
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - T. Alp Ikizler
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
- Vanderbilt Center for Immunobiology (VCI)
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4)
- Vanderbilt Institute for Global Health (VIGH)
| |
Collapse
|
28
|
Demirci M, Hinton A, Kirabo A. Dendritic cell epithelial sodium channel induced inflammation and salt-sensitive hypertension. Curr Opin Nephrol Hypertens 2024; 33:145-153. [PMID: 38180118 PMCID: PMC10842661 DOI: 10.1097/mnh.0000000000000963] [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] [Indexed: 01/06/2024]
Abstract
PURPOSE OF REVIEW Salt sensitivity of blood pressure (SSBP) is an independent risk factor for cardiovascular disease. Epithelial sodium channel (ENaC) plays a critical role in renal electrolyte and volume regulation and has been implicated in the pathogenesis of SSBP. This review describes recent advances regarding the role of ENaC-dependent inflammation in the development of SSBP. RECENT FINDINGS We recently found that sodium enters dendritic cells via ENaC, a process regulated by serum/glucocorticoid-regulated kinase 1 and epoxyeicosatrienoic acid 14,15. Sodium entry activates NADPH oxidase, leading to the production of isolevuglandins (IsoLGs). IsoLGs adduct self-proteins to form neoantigens in dendritic cells that activate T cells and result in the release of cytokines promoting sodium retention, kidney damage, and endothelial dysfunction in SSBP. Additionally, we described a novel mechanistic pathway involving ENaC and IsoLG-dependent NLRP3 inflammasome activation. These findings hold promise for the development of novel diagnostic biomarkers and therapeutic options for SSBP. SUMMARY The exact mechanisms underlying SSBP remain elusive. Recent advances in understanding the extrarenal role of ENaC have opened a new perspective, and further research efforts should focus on understanding the link between ENaC, inflammation, and SSBP.
Collapse
Affiliation(s)
- Mert Demirci
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health, Nashville, Tennessee, USA
| |
Collapse
|
29
|
Abstract
Salt (sodium chloride) is an essential nutrient required to maintain physiological functions. However, for most people, daily salt intake far exceeds their physiological need and is habitually greater than recommended upper thresholds. Excess salt intake leads to elevation in blood pressure which drives cardiovascular morbidity and mortality. Indeed, excessive salt intake is estimated to be responsible for ≈5 million deaths per year globally. For approximately one-third of otherwise healthy individuals (and >50% of those with hypertension), the effect of salt intake on blood pressure elevation is exaggerated; such people are categorized as salt sensitive and salt sensitivity of blood pressure is considered an independent risk factor for cardiovascular disease and death. The prevalence of salt sensitivity is higher in women than in men and, in both, increases with age. This narrative review considers the foundational concepts of salt sensitivity and the underlying effector systems that cause salt sensitivity. We also consider recent updates in preclinical and clinical research that are revealing new modifying factors that determine the blood pressure response to high salt intake.
Collapse
Affiliation(s)
- Matthew A Bailey
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom (M.A.B., N.D.)
| | - Neeraj Dhaun
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom (M.A.B., N.D.)
- Department of Renal Medicine, Royal Infirmary of Edinburgh, United Kingdom (N.D.)
| |
Collapse
|
30
|
Saleem M, Masenga SK, Ishimwe JA, Demirci M, Ahmad T, Jamison S, Albritton CF, Mwesigwa N, Porcia Haynes A, White J, Neikirk K, Vue Z, Hinton A, Arshad S, Desta S, Kirabo A. Recent Advances in Understanding Peripheral and Gut Immune Cell-Mediated Salt-Sensitive Hypertension and Nephropathy. Hypertension 2024; 81:436-446. [PMID: 38164753 PMCID: PMC10922672 DOI: 10.1161/hypertensionaha.123.22031] [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] [Indexed: 01/03/2024]
Abstract
Hypertension is the primary modifiable risk factor for cardiovascular, renal, and cerebrovascular diseases and is considered the main contributing factor to morbidity and mortality worldwide. Approximately 50% of hypertensive and 25% of normotensive people exhibit salt sensitivity of blood pressure, which is an independent risk factor for cardiovascular disease. Human and animal studies demonstrate that the immune system plays an important role in the etiology and pathogenesis of salt sensitivity of blood pressure, kidney damage, and vascular diseases. Antigen-presenting and adaptive immune cells are implicated in salt-sensitive hypertension and salt-induced renal and vascular injury. Elevated sodium activates antigen-presenting cells to release proinflammatory cytokines including IL (interleukin) 6, tumor necrosis factor-α, IL-1β, and accumulate isolevuglandin-protein adducts. In turn, these activate T cells release prohypertensive cytokines including IL-17A. Moreover, high-salt intake is associated with gut dysbiosis, leading to inflammation, oxidative stress, and blood pressure elevation but the mechanistic contribution to salt-sensitivity of blood pressure is not clearly understood. Here, we discuss recent advances in research investigating the cause, potential biomarkers, and therapeutic targets for salt-sensitive hypertension as they pertain to the gut microbiome, immunity, and inflammation.
Collapse
Affiliation(s)
- Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sepiso K Masenga
- Mulungushi University, School of Medicine and Health Sciences, HAND Research Group, Livingstone, Zambia
| | - Jeanne A Ishimwe
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mert Demirci
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Taseer Ahmad
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Punjab, Pakistan
| | - Sydney Jamison
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- School of Graduate Studies, Meharry Medical College, Nashville, TN, USA
| | - Claude F. Albritton
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- School of Graduate Studies, Meharry Medical College, Nashville, TN, USA
| | - Naome Mwesigwa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexandria Porcia Haynes
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jalyn White
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Spelman College Department of Chemistry and Biochemistry, Atlanta, GA, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Suha Arshad
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Selam Desta
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health
| |
Collapse
|
31
|
Nosalski R, Siedlinski M, Neves KB, Monaco C. Editorial: The interplay between oxidative stress, immune cells and inflammation in cardiovascular diseases. Front Cardiovasc Med 2024; 11:1385809. [PMID: 38476378 PMCID: PMC10928531 DOI: 10.3389/fcvm.2024.1385809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Affiliation(s)
- R. Nosalski
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - M. Siedlinski
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Omicron Medical Genomics Laboratory, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - K. B. Neves
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde Glasgow, Glasgow, United Kingdom
| | - C. Monaco
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
32
|
Zhang H, Dhalla NS. The Role of Pro-Inflammatory Cytokines in the Pathogenesis of Cardiovascular Disease. Int J Mol Sci 2024; 25:1082. [PMID: 38256155 PMCID: PMC10817020 DOI: 10.3390/ijms25021082] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
With cardiovascular disease (CVD) being a primary source of global morbidity and mortality, it is crucial that we understand the molecular pathophysiological mechanisms at play. Recently, numerous pro-inflammatory cytokines have been linked to several different CVDs, which are now often considered an adversely pro-inflammatory state. These cytokines most notably include interleukin-6 (IL-6),tumor necrosis factor (TNF)α, and the interleukin-1 (IL-1) family, amongst others. Not only does inflammation have intricate and complex interactions with pathophysiological processes such as oxidative stress and calcium mishandling, but it also plays a role in the balance between tissue repair and destruction. In this regard, pre-clinical and clinical evidence has clearly demonstrated the involvement and dynamic nature of pro-inflammatory cytokines in many heart conditions; however, the clinical utility of the findings so far remains unclear. Whether these cytokines can serve as markers or risk predictors of disease states or act as potential therapeutic targets, further extensive research is needed to fully understand the complex network of interactions that these molecules encompass in the context of heart disease. This review will highlight the significant advances in our understanding of the contributions of pro-inflammatory cytokines in CVDs, including ischemic heart disease (atherosclerosis, thrombosis, acute myocardial infarction, and ischemia-reperfusion injury), cardiac remodeling (hypertension, cardiac hypertrophy, cardiac fibrosis, cardiac apoptosis, and heart failure), different cardiomyopathies as well as ventricular arrhythmias and atrial fibrillation. In addition, this article is focused on discussing the shortcomings in both pathological and therapeutic aspects of pro-inflammatory cytokines in CVD that still need to be addressed by future studies.
Collapse
Affiliation(s)
- Hannah Zhang
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
33
|
Haynes AP, Desta S, Ahmad T, Neikirk K, Hinton A, Bloodworth N, Kirabo A. The Antioxidative Effects of Flavones in Hypertensive Disease. Biomedicines 2023; 11:2877. [PMID: 38001878 PMCID: PMC10669108 DOI: 10.3390/biomedicines11112877] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 11/26/2023] Open
Abstract
Hypertension is the leading remediable risk factor for cardiovascular morbidity and mortality in the United States. Excess dietary salt consumption, which is a catalyst of hypertension, initiates an inflammatory cascade via activation of antigen-presenting cells (APCs). This pro-inflammatory response is driven primarily by sodium ions (Na+) transporting into APCs by the epithelial sodium channel (ENaC) and subsequent NADPH oxidase activation, leading to high levels of oxidative stress. Oxidative stress, a well-known catalyst for hypertension-related illness development, disturbs redox homeostasis, which ultimately promotes lipid peroxidation, isolevuglandin production and an inflammatory response. Natural medicinal compounds derived from organic materials that are characterized by their anti-inflammatory, anti-oxidative, and anti-mutagenic properties have recently gained traction amongst the pharmacology community due to their therapeutic effects. Flavonoids, a natural phenolic compound, have these therapeutic benefits and can potentially serve as anti-hypertensives. Flavones are a type of flavonoid that have increased anti-inflammatory effects that may allow them to act as therapeutic agents for hypertension, including diosmetin, which is able to induce significant arterial vasodilation in several different animal models. This review will focus on the activity of flavones to illuminate potential preventative and potential therapeutic mechanisms against hypertension.
Collapse
Affiliation(s)
- Alexandria Porcia Haynes
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN 37212, USA; (A.P.H.); (S.D.); (T.A.)
| | - Selam Desta
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN 37212, USA; (A.P.H.); (S.D.); (T.A.)
- Department of Biology, College of Arts and Sciences, Howard University, Washington, DC 20059, USA
| | - Taseer Ahmad
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN 37212, USA; (A.P.H.); (S.D.); (T.A.)
- Department of Pharmacology, College of Pharmacy, University of Sargodha, University Road, Sargodha 40100, Punjab, Pakistan
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN 37235, USA; (K.N.); (A.H.)
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN 37235, USA; (K.N.); (A.H.)
| | - Nathaniel Bloodworth
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN 37212, USA; (A.P.H.); (S.D.); (T.A.)
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN 37212, USA; (A.P.H.); (S.D.); (T.A.)
| |
Collapse
|
34
|
Wu Q, Meng W, Zhu B, Chen X, Fu J, Zhao C, Liu G, Luo X, Lv Y, Zhao W, Wang F, Hu S, Zhang S. VEGFC ameliorates salt-sensitive hypertension and hypertensive nephropathy by inhibiting NLRP3 inflammasome via activating VEGFR3-AMPK dependent autophagy pathway. Cell Mol Life Sci 2023; 80:327. [PMID: 37837447 PMCID: PMC11072217 DOI: 10.1007/s00018-023-04978-3] [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: 05/25/2023] [Revised: 08/29/2023] [Accepted: 09/23/2023] [Indexed: 10/16/2023]
Abstract
Salt-sensitivity hypertension (SSHTN) is an independent predictor for cardiovascular mortality. VEGFC has been reported to be a protective role in SSHTN and hypertensive kidney injury. However, the underlying mechanisms remain largely unclear. The current study aimed to explore the protective effects and mechanisms of VEGFC against SSHTN and hypertensive nephropathy. Here, we reported that VEGFC attenuated high blood pressure as well as protected against renal inflammation and fibrosis in SSHTN mice. Moreover, VEGFC suppressed the activation of renal NLRP3 inflammasome in SSHTN mice. In vitro, we found VEGFC inhibited NLRP3 inflammasome activation, meanwhile, upregulated autophagy in high-salt-induced macrophages, while these effects were reversed by an autophagy inhibitor 3MA. Furthermore, in vivo, 3MA pretreatment weakened the protective effects of VEGFC on SSHTN and hypertensive nephropathy. Mechanistically, VEGF receptor 3 (VEGFR3) kinase domain activated AMPK by promoting the phosphorylation at Thr183 via binding to AMPK, thus enhancing autophagy activity in the context of high-salt-induced macrophages. These findings indicated that VEGFC inhibited NLRP3 inflammasome activation by promoting VEGFR3-AMPK-dependent autophagy pathway in high-salt-induced macrophages, which provided a mechanistic basis for the therapeutic target in SSHTN and hypertensive kidney injury.
Collapse
Affiliation(s)
- Qiuwen Wu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Wei Meng
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Bin Zhu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Xi Chen
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Jiaxin Fu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Chunyu Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Gang Liu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xing Luo
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Ying Lv
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Wenqi Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Fan Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Sining Hu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China.
| | - Shuo Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
35
|
Jang S, Kim JY, Kim CW, Kim I. Hypertonic Salt Solution Enhances Inflammatory Responses in Cultured Splenic T-Cells from Dahl Salt-Sensitive Rats but Not Dahl Salt-Resistant Rats. J Cardiovasc Dev Dis 2023; 10:414. [PMID: 37887861 PMCID: PMC10607114 DOI: 10.3390/jcdd10100414] [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: 08/07/2023] [Revised: 09/12/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
This study aimed to delineate the effect of sodium chloride on the induction of inflammatory responses and the development of hypertension in Dahl salt-sensitive (SS) and salt-resistant (SR) rats. Splenocytes were isolated from the spleens of SS and SR rats, and cultured on anti-CD3-coated plates for 5 days. The cultured splenic T-cells were challenged with a hypertonic salt solution (0, 20, or 40 mM) in the absence or presence of IL-6 (0, 20, or 60 ng/mL), TGF-β (0, 5, or 15 ng/mL), or IL-23 (0, 10, or 30 ng/mL), and analyzed via ELISA, flow cytometry, and immunofluorescence. The hypertonic salt solution potentiated IL-17A production, as well as the differentiation of Th17 cells via IL-6/TGF-β/IL-23, exclusively in SS rats. However, it did not affect IL-10 production or the differentiation of Treg cells in any of the groups. Furthermore, it potentiated the signal of RORγt in IL-6-treated splenic T-cells from SS rats. To summarize, cultured splenic T-cells exhibited enhanced inflammatory responses on exposure to a hypertonic salt solution in SS rats only, which indicated that sodium chloride and inflammatory cytokines synergistically drove the induction of pathogenic Th17 cells and the development of hypertension in this group only.
Collapse
Affiliation(s)
- Sungmin Jang
- Department of Pharmacology, School of Medicine, Daegu, 41944, Republic of Korea; (S.J.); (J.Y.K.); (C.-W.K.)
- Cardiovascular Research Institute, Daegu 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Daegu 41944, Republic of Korea
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Jee Young Kim
- Department of Pharmacology, School of Medicine, Daegu, 41944, Republic of Korea; (S.J.); (J.Y.K.); (C.-W.K.)
- Cardiovascular Research Institute, Daegu 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Daegu 41944, Republic of Korea
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Cheong-Wun Kim
- Department of Pharmacology, School of Medicine, Daegu, 41944, Republic of Korea; (S.J.); (J.Y.K.); (C.-W.K.)
- Cardiovascular Research Institute, Daegu 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Daegu 41944, Republic of Korea
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Inkyeom Kim
- Department of Pharmacology, School of Medicine, Daegu, 41944, Republic of Korea; (S.J.); (J.Y.K.); (C.-W.K.)
- Cardiovascular Research Institute, Daegu 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Daegu 41944, Republic of Korea
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
36
|
Qi J, Wang J, Zhang Y, Long H, Dong L, Wan P, Zuo Z, Chen W, Song Z. High-Salt-Diet (HSD) aggravates the progression of Inflammatory Bowel Disease (IBD) via regulating epithelial necroptosis. MOLECULAR BIOMEDICINE 2023; 4:28. [PMID: 37691056 PMCID: PMC10493205 DOI: 10.1186/s43556-023-00135-1] [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: 12/29/2022] [Accepted: 06/14/2023] [Indexed: 09/12/2023] Open
Abstract
Due to its unclear etiology, there is no specific medicine to cure the recurrent and incurable inflammatory bowel disease (IBD). Unhealthy dietary habits unconsciously contributed to the progression of IBD, for example a High-Salt-Diet (HSD) is the most neglected and frequently adopted habit. However, the molecular mechanism of how HSD aggravates the progression of IBD has yet to remain uncovered. Herein, we focus on the hypothesis that necroptosis pathway may be involved in the process of IBD exacerbated by HSD. To this end, different gene expression (DEGs) profiles of human epithelia under hypertonic culture conditions were applied to screen candidate pathways. What's more, gene expression manipulation, immune microenvironment detection, RIPK3/MLKL gene knockout (KO), and wild-type (WT) mice were carried out to research the promotion of IBD progression under treatments of high salt intake. Based on our present results, gene expression profiles in human normal colon epithelia cell NCM460 were significantly changed under salt- or sucrose-induced hypertonic culture conditions. RIPK3 was significantly up-regulated under both conditions. Furthermore, mice colon epithelia cell CT26 growth was inhibited in a time- and dose-dependent manner by extra NaCl incubation. Autophagy, and Necroptosis pathways were activated and enhanced by LPS pretreatment. HSD significantly exacerbated DSS-induced IBD symptoms in vivo in a dose-dependent manner. Moreover, RIPK3-/- and MLKL-/- mice presented severe IBD symptoms in vivo. Overall, the results demonstrated that HSD aggravated the IBD progression via necroptosis activation, providing novel strategies and promising targets for the clinical treatment of IBD.
Collapse
Affiliation(s)
- Jialong Qi
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
- Yunnan Provincial Key Laboratory of Clinical Virology, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
| | - Jinli Wang
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
| | - Ying Zhang
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
- School of Medicine, Kunming University of Science and Technology, Affiliated By The First People's Hospital of Yunnan Province, Kunming, 650504, Yunnan, P.R. China
| | - Huan Long
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
- School of Medicine, Kunming University of Science and Technology, Affiliated By The First People's Hospital of Yunnan Province, Kunming, 650504, Yunnan, P.R. China
| | - Liang Dong
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
- School of Medicine, Kunming University of Science and Technology, Affiliated By The First People's Hospital of Yunnan Province, Kunming, 650504, Yunnan, P.R. China
| | - Ping Wan
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
- School of Medicine, Kunming University of Science and Technology, Affiliated By The First People's Hospital of Yunnan Province, Kunming, 650504, Yunnan, P.R. China
| | - Zan Zuo
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China.
| | - Wenjie Chen
- State Key Laboratory of Respiratory Disease, Guangdong-Hongkong-Macao Joint Laboratory of Respiratory Infectious Disease, Guangzhou Medical University, Guangzhou, 510182, P.R. China.
- Sydney Vital Translational Cancer Research Centre, Westbourne St, Sydney, NSW, 2065, Australia.
| | - Zhengji Song
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China.
| |
Collapse
|
37
|
Zhuang T, Lei Y, Chang JJ, Zhou YP, Li Y, Li YX, Yang YF, Chen MH, Meng T, Fu SM, Huang LH, Cheang WS, Cooke JP, Dong ZH, Bai YN, Ruan CC. A2AR-mediated lymphangiogenesis via VEGFR2 signaling prevents salt-sensitive hypertension. Eur Heart J 2023; 44:2730-2742. [PMID: 37377160 PMCID: PMC10393074 DOI: 10.1093/eurheartj/ehad377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 04/17/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
AIMS Excess dietary sodium intake and retention lead to hypertension. Impaired dermal lymphangiogenesis and lymphatic dysfunction-mediated sodium and fluid imbalance are pathological mechanisms. The adenosine A2A receptor (A2AR) is expressed in lymphatic endothelial cells (LECs), while the roles and mechanisms of LEC-A2AR in skin lymphangiogenesis during salt-induced hypertension are not clear. METHODS AND RESULTS The expression of LEC-A2AR correlated with lymphatic vessel density in both high-salt diet (HSD)-induced hypertensive mice and hypertensive patients. Lymphatic endothelial cell-specific A2AR knockout mice fed HSD exhibited 17 ± 2% increase in blood pressure and 17 ± 3% increase in Na+ content associated with decreased lymphatic density (-19 ± 2%) compared with HSD-WT mice. A2AR activation by agonist CGS21680 increased lymphatic capillary density and decreased blood pressure in HSD-WT mice. Furthermore, this A2AR agonist activated MSK1 directly to promote VEGFR2 activation and endocytosis independently of VEGF as assessed by phosphoprotein profiling and immunoprecipitation assays in LECs. VEGFR2 kinase activity inhibitor fruquintinib or VEGFR2 knockout in LECs but not VEGF-neutralizing antibody bevacizumab suppressed A2AR activation-mediated decrease in blood pressure. Immunostaining revealed phosphorylated VEGFR2 and MSK1 expression in the LECs were positively correlated with skin lymphatic vessel density and A2AR level in hypertensive patients. CONCLUSION The study highlights a novel A2AR-mediated VEGF-independent activation of VEGFR2 signaling in dermal lymphangiogenesis and sodium balance, which might be a potential therapeutic target in salt-sensitive hypertension.
Collapse
Affiliation(s)
- Tao Zhuang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Yu Lei
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Jin-Jia Chang
- Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Yan-Ping Zhou
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pu-Jian Road, Shanghai 200032, China
| | - Yan Li
- Department of Cardiology, RuiJin Hospital/LuWan Branch, Shanghai Jiao Tong University School of Medicine, 149 Chong-Qing-Nan Road, Shanghai 200032, China
| | - Yan-Xiu Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guang-Zhou Road, Nanjing 210000, China
| | - Yong-Feng Yang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Mei-Hua Chen
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Ting Meng
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Shi-Man Fu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Li-Hao Huang
- Department of Chemistry and Institute of Metabolism and Integrative Biology, Shanghai Key Laboratory of Metabolic Remodeling and Health, Fudan University, 38 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Wai-San Cheang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Long-Ma Road, Macau 999078, China
| | - John P Cooke
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Zhi-Hui Dong
- Department of Vascular Surgery, Zhongshan Hospital, and Center for Vascular Surgery and Wound Care, Jinshan Hospital, Fudan University, 180 Feng-Lin Road, Shanghai 200032, China
| | - Ying-Nan Bai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Feng-Lin Road, Shanghai 200032, China
| | - Cheng-Chao Ruan
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| |
Collapse
|
38
|
Chen Y, Yu X, Yan Z, Zhang S, Zhang J, Guo W. Role of epithelial sodium channel-related inflammation in human diseases. Front Immunol 2023; 14:1178410. [PMID: 37559717 PMCID: PMC10407551 DOI: 10.3389/fimmu.2023.1178410] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/29/2023] [Indexed: 08/11/2023] Open
Abstract
The epithelial sodium channel (ENaC) is a heterotrimer and is widely distributed throughout the kidneys, blood vessels, lungs, colons, and many other organs. The basic role of the ENaC is to mediate the entry of Na+ into cells; the ENaC also has an important regulatory function in blood pressure, airway surface liquid (ASL), and endothelial cell function. Aldosterone, serum/glucocorticoid kinase 1 (SGK1), shear stress, and posttranslational modifications can regulate the activity of the ENaC; some ion channels also interact with the ENaC. In recent years, it has been found that the ENaC can lead to immune cell activation, endothelial cell dysfunction, aggravated inflammation involved in high salt-induced hypertension, cystic fibrosis, pseudohypoaldosteronism (PHA), and tumors; some inflammatory cytokines have been reported to have a regulatory role on the ENaC. The ENaC hyperfunction mediates the increase of intracellular Na+, and the elevated exchange of Na+ with Ca2+ leads to an intracellular calcium overload, which is an important mechanism for ENaC-related inflammation. Some of the research on the ENaC is controversial or unclear; we therefore reviewed the progress of studies on the role of ENaC-related inflammation in human diseases and their mechanisms.
Collapse
Affiliation(s)
- Yabin Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- National Organ Transplantation (Liver &Kidney Transplantation) Physician Training Centre, Zhengzhou, China
- National Regional Medical Treatment Centre of Henan Organ Transplantation, Zhengzhou, China
| | - Xiao Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- National Organ Transplantation (Liver &Kidney Transplantation) Physician Training Centre, Zhengzhou, China
- National Regional Medical Treatment Centre of Henan Organ Transplantation, Zhengzhou, China
| | - Zhiping Yan
- Henan Organ Transplantation Centre, Zhengzhou, China
- Henan Engineering and Research Center for Diagnosis and Treatment of Hepatobiliary and Pancreatic Surgical Diseases, Zhengzhou, China
| | - Shuijun Zhang
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
| | - Jiacheng Zhang
- Henan Key Laboratory for Digestive Organ Transplantation, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Open and Key Laboratory for Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, China
| |
Collapse
|
39
|
Ertuglu LA, Pitzer Mutchler A, Elijovich F, Laffer CL, Sheng Q, Wanjalla CN, Kirabo A. Regulation of human salt-sensitivite hypertension by myeloid cell renin-angiotensin-aldosterone system. Front Physiol 2023; 14:1208270. [PMID: 37534363 PMCID: PMC10390697 DOI: 10.3389/fphys.2023.1208270] [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: 04/18/2023] [Accepted: 06/06/2023] [Indexed: 08/04/2023] Open
Abstract
Introduction: Salt sensitivity of blood pressure is a phenomenon in which blood pressure changes according to dietary sodium intake. Our previous studies found that high salt activates antigen presenting cells, resulting in the development of hypertension. The mechanisms by which salt-induced immune cell activation is regulated in salt sensitivity of blood pressure are unknown. In the current study, we investigated dietary salt-induced effects on the renin-angiotensin-aldosterone system (RAAS) gene expression in myeloid immune cells and their impact on salt sensitive hypertension in humans. Methods: We performed both bulk and single-cell sequencing analysis on immune cells with in vitro and in vivo high dietary salt treatment in humans using a rigorous salt-loading/depletion protocol to phenotype salt-sensitivity of blood pressure. We also measured plasma renin and aldosterone using radioimmunoassay. Results: We found that while in vitro high sodium exposure downregulated the expression of renin, renin binding protein and renin receptor, there were no significant changes in the genes of the renin-angiotensin system in response to dietary salt loading and depletion in vivo. Plasma renin in salt sensitive individuals tended to be lower with a blunted response to the salt loading/depletion challenge as previously reported. Discussion: These findings suggest that unlike systemic RAAS, acute changes in dietary salt intake do not regulate RAAS expression in myeloid immune cells.
Collapse
Affiliation(s)
- Lale A. Ertuglu
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ashley Pitzer Mutchler
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, Nashville, TN, United States
| | - Fernando Elijovich
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, Nashville, TN, United States
| | - Cheryl L. Laffer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, Nashville, TN, United States
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Celestine N. Wanjalla
- Department of Internal Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center Nashville, Nashville, TN, United States
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, Nashville, TN, United States
| |
Collapse
|
40
|
Sembajwe LF, Ssekandi AM, Namaganda A, Muwonge H, Kasolo JN, Kalyesubula R, Nakimuli A, Naome M, Patel KP, Masenga SK, Kirabo A. Glycocalyx-Sodium Interaction in Vascular Endothelium. Nutrients 2023; 15:2873. [PMID: 37447199 PMCID: PMC10343370 DOI: 10.3390/nu15132873] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
The glycocalyx generally covers almost all cellular surfaces, where it participates in mediating cell-surface interactions with the extracellular matrix as well as with intracellular signaling molecules. The endothelial glycocalyx that covers the luminal surface mediates the interactions of endothelial cells with materials flowing in the circulating blood, including blood cells. Cardiovascular diseases (CVD) remain a major cause of morbidity and mortality around the world. The cardiovascular risk factors start by causing endothelial cell dysfunction associated with destruction or irregular maintenance of the glycocalyx, which may culminate into a full-blown cardiovascular disease. The endothelial glycocalyx plays a crucial role in shielding the cell from excessive exposure and absorption of excessive salt, which can potentially cause damage to the endothelial cells and underlying tissues of the blood vessels. So, in this mini review/commentary, we delineate and provide a concise summary of the various components of the glycocalyx, their interaction with salt, and subsequent involvement in the cardiovascular disease process. We also highlight the major components of the glycocalyx that could be used as disease biomarkers or as drug targets in the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Lawrence Fred Sembajwe
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Abdul M. Ssekandi
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Agnes Namaganda
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Haruna Muwonge
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Josephine N. Kasolo
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Robert Kalyesubula
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Annettee Nakimuli
- Department of Obstetrics and Gynecology, School of Medicine, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda;
| | - Mwesigwa Naome
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Sepiso K. Masenga
- Department of Physiological Sciences, School of Medicine and Health Sciences, Mulungushi University, Kabwe P.O. Box 80415, Zambia;
| | - Annet Kirabo
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| |
Collapse
|
41
|
Masenga SK, Kirabo A. Hypertensive heart disease: risk factors, complications and mechanisms. Front Cardiovasc Med 2023; 10:1205475. [PMID: 37342440 PMCID: PMC10277698 DOI: 10.3389/fcvm.2023.1205475] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/26/2023] [Indexed: 06/22/2023] Open
Abstract
Hypertensive heart disease constitutes functional and structural dysfunction and pathogenesis occurring primarily in the left ventricle, the left atrium and the coronary arteries due to chronic uncontrolled hypertension. Hypertensive heart disease is underreported and the mechanisms underlying its correlates and complications are not well elaborated. In this review, we summarize the current understanding of hypertensive heart disease, we discuss in detail the mechanisms associated with development and complications of hypertensive heart disease especially left ventricular hypertrophy, atrial fibrillation, heart failure and coronary artery disease. We also briefly highlight the role of dietary salt, immunity and genetic predisposition in hypertensive heart disease pathogenesis.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Cam-Pus, Livingstone, Zambia
- School of Medicine, University of Zambia, Lusaka, Zambia
- Department of Medicine, Vanderbilt University Medical Centre, Nashville, TN, United States
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Centre, Nashville, TN, United States
| |
Collapse
|
42
|
Abstract
Epidemiologic studies detected an inverse relationship between HDL (high-density lipoprotein) cholesterol (HDL-C) levels and atherosclerotic cardiovascular disease (ASCVD), identifying HDL-C as a major risk factor for ASCVD and suggesting atheroprotective functions of HDL. However, the role of HDL-C as a mediator of risk for ASCVD has been called into question by the failure of HDL-C-raising drugs to reduce cardiovascular events in clinical trials. Progress in understanding the heterogeneous nature of HDL particles in terms of their protein, lipid, and small RNA composition has contributed to the realization that HDL-C levels do not necessarily reflect HDL function. The most examined atheroprotective function of HDL is reverse cholesterol transport, whereby HDL removes cholesterol from plaque macrophage foam cells and delivers it to the liver for processing and excretion into bile. Indeed, in several studies, HDL has shown inverse associations between HDL cholesterol efflux capacity and ASCVD in humans. Inflammation plays a key role in the pathogenesis of atherosclerosis and vulnerable plaque formation, and a fundamental function of HDL is suppression of inflammatory signaling in macrophages and other cells. Oxidation is also a critical process to ASCVD in promoting atherogenic oxidative modifications of LDL (low-density lipoprotein) and cellular inflammation. HDL and its proteins including apoAI (apolipoprotein AI) and PON1 (paraoxonase 1) prevent cellular oxidative stress and LDL modifications. Importantly, HDL in humans with ASCVD is oxidatively modified rendering HDL dysfunctional and proinflammatory. Modification of HDL with reactive carbonyl species, such as malondialdehyde and isolevuglandins, dramatically impairs the antiatherogenic functions of HDL. Importantly, treatment of murine models of atherosclerosis with scavengers of reactive dicarbonyls improves HDL function and reduces systemic inflammation, atherosclerosis development, and features of plaque instability. Here, we discuss the HDL antiatherogenic functions in relation to oxidative modifications and the potential of reactive dicarbonyl scavengers as a therapeutic approach for ASCVD.
Collapse
Affiliation(s)
- MacRae F. Linton
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
- 2. Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Patricia G. Yancey
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Huan Tao
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Sean S. Davies
- 2. Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
43
|
Pitzer Mutchler A, Huynh L, Patel R, Lam T, Bain D, Jamison S, Kirabo A, Ray EC. The role of dietary magnesium deficiency in inflammatory hypertension. Front Physiol 2023; 14:1167904. [PMID: 37293263 PMCID: PMC10244581 DOI: 10.3389/fphys.2023.1167904] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/10/2023] [Indexed: 06/10/2023] Open
Abstract
Nearly 30% of adults consume less than the estimated average daily requirement of magnesium (Mg2+), and commonly used medications, such as diuretics, promote Mg2+ deficiency. Higher serum Mg2+ levels, increased dietary Mg2+ in-take, and Mg2+ supplementation are each associated with lower blood pressure, suggesting that Mg2+-deficiency contributes to the pathogenesis of hypertension. Antigen-presenting cells, such as monocytes and dendritic cells, are well-known to be involved in the pathogenesis of hypertension. In these cells, processes implicated as necessary for increased blood pressure include activation of the NLRP3 inflammasome, IL-1β production, and oxidative modification of fatty acids such as arachidonic acid, forming isolevuglandins (IsoLGs). We hypothesized that increased blood pressure in response to dietary Mg2+-depletion leads to increased NLRP3, IL-1β, and IsoLG production in antigen presenting cells. We found that a Mg2+-depleted diet (0.01% Mg2+ diet) increased blood pressure in mice compared to mice fed a 0.08% Mg2+ diet. Mg2+-depleted mice did not exhibit an increase in total body fluid, as measured by quantitative magnetic resonance. Plasma IL-1β concentrations were increased (0.13 ± 0.02 pg/mL vs. 0.04 ± 0.02 pg/mL). Using flow cytometry, we observed increased NLRP3 and IL-1β expression in antigen-presenting cells from spleen, kidney, and aorta. We also observed increased IsoLG production in antigen-presenting cells from these organs. Primary culture of CD11c+ dendritic cells confirmed that low extracellular Mg2+ exerts a direct effect on these cells, stimulating IL-1β and IL-18 production. The present findings show that NLRP3 inflammasome activation and IsoLG-adduct formation are stimulated when dietary Mg2+ is depleted. Interventions and increased dietary Mg2+ consumption may prove beneficial in decreasing the prevalence of hypertension and cardiovascular disease.
Collapse
Affiliation(s)
- Ashley Pitzer Mutchler
- Vanderbilt University Department of Medicine, Division of Clinical Pharmacology, Nashville, TN, United States
| | - Linh Huynh
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| | - Ritam Patel
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| | - Tracey Lam
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| | - Daniel Bain
- University of Pittsburgh Department of Geology, Pittsburgh, PA, United States
| | - Sydney Jamison
- Meharry Medical College Nashville, Nashville, TN, United States
| | - Annet Kirabo
- Vanderbilt University Department of Medicine, Division of Clinical Pharmacology, Nashville, TN, United States
| | - Evan C. Ray
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| |
Collapse
|
44
|
Abstract
Hypertension is a major healthcare issue that afflicts one in every three adults worldwide and contributes to cardiovascular diseases, morbidity and mortality. Bioactive lipids contribute importantly to blood pressure regulation via actions on the vasculature, kidney, and inflammation. Vascular actions of bioactive lipids include blood pressure lowering vasodilation and blood pressure elevating vasoconstriction. Increased renin release by bioactive lipids in the kidney is pro-hypertensive whereas anti-hypertensive bioactive lipid actions result in increased sodium excretion. Bioactive lipids have pro-inflammatory and anti-inflammatory actions that increase or decrease reactive oxygen species and impact vascular and kidney function in hypertension. Human studies provide evidence that fatty acid metabolism and bioactive lipids contribute to sodium and blood pressure regulation in hypertension. Genetic changes identified in humans that impact arachidonic acid metabolism have been associated with hypertension. Arachidonic acid cyclooxygenase, lipoxygenase and cytochrome P450 metabolites have pro-hypertensive and anti-hypertensive actions. Omega-3 fish oil fatty acids eicosapentaenoic acid and docosahexaenoic acid are known to be anti-hypertensive and cardiovascular protective. Lastly, emerging fatty acid research areas include blood pressure regulation by isolevuglandins, nitrated fatty acids, and short chain fatty acids. Taken together, bioactive lipids are key contributors to blood pressure regulation and hypertension and their manipulation could decrease cardiovascular disease and associated morbidity and mortality.
Collapse
Affiliation(s)
- John D Imig
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
45
|
Ma J, Li Y, Yang X, Liu K, Zhang X, Zuo X, Ye R, Wang Z, Shi R, Meng Q, Chen X. Signaling pathways in vascular function and hypertension: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:168. [PMID: 37080965 PMCID: PMC10119183 DOI: 10.1038/s41392-023-01430-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
Hypertension is a global public health issue and the leading cause of premature death in humans. Despite more than a century of research, hypertension remains difficult to cure due to its complex mechanisms involving multiple interactive factors and our limited understanding of it. Hypertension is a condition that is named after its clinical features. Vascular function is a factor that affects blood pressure directly, and it is a main strategy for clinically controlling BP to regulate constriction/relaxation function of blood vessels. Vascular elasticity, caliber, and reactivity are all characteristic indicators reflecting vascular function. Blood vessels are composed of three distinct layers, out of which the endothelial cells in intima and the smooth muscle cells in media are the main performers of vascular function. The alterations in signaling pathways in these cells are the key molecular mechanisms underlying vascular dysfunction and hypertension development. In this manuscript, we will comprehensively review the signaling pathways involved in vascular function regulation and hypertension progression, including calcium pathway, NO-NOsGC-cGMP pathway, various vascular remodeling pathways and some important upstream pathways such as renin-angiotensin-aldosterone system, oxidative stress-related signaling pathway, immunity/inflammation pathway, etc. Meanwhile, we will also summarize the treatment methods of hypertension that targets vascular function regulation and discuss the possibility of these signaling pathways being applied to clinical work.
Collapse
Affiliation(s)
- Jun Ma
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yanan Li
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiangyu Yang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xin Zhang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xianghao Zuo
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Runyu Ye
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ziqiong Wang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Rufeng Shi
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Qingtao Meng
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
46
|
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
|
47
|
Benson LN, Guo Y, Deck K, Mora C, Liu Y, Mu S. The link between immunity and hypertension in the kidney and heart. Front Cardiovasc Med 2023; 10:1129384. [PMID: 36970367 PMCID: PMC10034415 DOI: 10.3389/fcvm.2023.1129384] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
Hypertension is the primary cause of cardiovascular disease, which is a leading killer worldwide. Despite the prevalence of this non-communicable disease, still between 90% and 95% of cases are of unknown or multivariate cause ("essential hypertension"). Current therapeutic options focus primarily on lowering blood pressure through decreasing peripheral resistance or reducing fluid volume, but fewer than half of hypertensive patients can reach blood pressure control. Hence, identifying unknown mechanisms causing essential hypertension and designing new treatment accordingly are critically needed for improving public health. In recent years, the immune system has been increasingly implicated in contributing to a plethora of cardiovascular diseases. Many studies have demonstrated the critical role of the immune system in the pathogenesis of hypertension, particularly through pro-inflammatory mechanisms within the kidney and heart, which, eventually, drive a myriad of renal and cardiovascular diseases. However, the precise mechanisms and potential therapeutic targets remain largely unknown. Therefore, identifying which immune players are contributing to local inflammation and characterizing pro-inflammatory molecules and mechanisms involved will provide promising new therapeutic targets that could lower blood pressure and prevent progression from hypertension into renal or cardiac dysfunction.
Collapse
Affiliation(s)
- Lance N. Benson
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, United States
| | | | | | | | | | - Shengyu Mu
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, United States
| |
Collapse
|
48
|
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
|
49
|
Ishimwe JA, Ferguson JF, Kirabo A. Sex Differences in Fatty Acid Metabolism and Blood Pressure Response to Dietary Salt in Humans. CARDIOGENETICS 2023; 13:33-46. [PMID: 38605973 PMCID: PMC11008634 DOI: 10.3390/cardiogenetics13010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
Salt sensitivity is a trait in which high dietary sodium (Na+) intake causes an increase in blood pressure (BP). We previously demonstrated that in the gut, elevated dietary Na+ causes dysbiosis. The mechanistic interplay between excess dietary Na+-induced alteration in the gut microbiome and sex differences is less understood. The goal of this study was to identify novel metabolites in sex differences and blood pressure in response to a high dietary Na+ intake. We performed stool and plasma metabolomics analysis and measured the BP of human volunteers with salt intake above or below the American Heart Association recommendations. We also performed RNA sequencing on human monocytes treated with high salt in vitro. The relationship between BP and dietary Na+ intake was different in women and men. Network analysis revealed that fatty acids as top subnetworks differentially changed with salt intake. We found that women with high dietary Na+ intake have high levels of arachidonic acid related metabolism, suggesting a role in sex differences of the blood pressure response to Na+. The exposure of monocytes to high salt in vitro upregulates the transcription of fatty acid receptors and arachidonic acid-related genes. These findings provide potentially novel insights into metabolic changes underlying gut dysbiosis and inflammation in salt sensitivity of BP.
Collapse
Affiliation(s)
- Jeanne A. Ishimwe
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
| | - Jane F. Ferguson
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Nashville, TN 37235, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Medical Center, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Hypertension remains a global health and socioeconomic burden. Immune mechanisms are now recognized as integral part of the multifactorial etiology of hypertension and related organ damage. The present review addresses inflammatory pathways and immune targets in hypertension, which may be important for an immunomodulatory treatment of hypertension aside from lowering arterial pressure. RECENT FINDINGS Anti-inflammatory interventions targeting single interleukins or almost the entire immune system show different beneficial effects. While immunomodulation (targeting specific portion of immune system) shows beneficial outcomes in certain groups of hypertensives, this does not pertain to immunosuppression (targeting entire immune system). Immunomodulatory interventions improve outcomes of hypertension independent of arterial pressure. The studies reveal interleukins, such as interleukin (IL)-1β and IL-17 as targets of immunomodulation. Besides interleukins, targeting αvβ-3 integrin and matrix metalloproteinase-2 or using experimental cell-therapy demonstrate beneficial effects in hypertensive organ damage. The NLR family pyrin domain containing 3 (NLRP3) inflammasome/IL-1β/endothelial cell/T-cell axis seems to be an important mediator in sustained inflammation during hypertension. SUMMARY Although immunomodulation may be advantageous as a causal therapy in hypertension, targeting immune networks rather than single interleukins appears of major importance. Further research is required to better identify these networks and their links to human hypertension.
Collapse
Affiliation(s)
- Andreas Deussen
- Department of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | |
Collapse
|