1
|
Shi MQ, Chen J, Ji FH, Zhou H, Peng K, Wang J, Fan CL, Wang X, Wang Y. Prognostic impact of hypernatremia for septic shock patients in the intensive care unit. World J Clin Cases 2025; 13:95430. [PMID: 40051797 PMCID: PMC11612684 DOI: 10.12998/wjcc.v13.i7.95430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 10/04/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Hypernatremia represents a significant electrolyte imbalance associated with numerous adverse outcomes, particularly in cases of intensive care unit (ICU)-acquired hypernatremia (IAH). Nevertheless, its relevance in patients with septic shock remains uncertain. AIM To identify independent risk factors and their predictive efficacy for IAH to improve outcomes in patients with septic shock. METHODS In the present retrospective single-center study, a cohort of 157 septic shock patients with concurrent hypernatremia in the ICU at The First Affiliated Hospital of Soochow University, between August 1, 2018, and May 31, 2023, were analyzed. Patients were categorized based on the timing of hypernatremia occurrence into the IAH group (n = 62), the non-IAH group (n = 41), and the normonatremia group (n = 54). RESULTS In the present study, there was a significant association between the high serum sodium concentrations, excessive persistent inflammation, immunosuppression and catabolism syndrome and chronic critical illness, while rapid recovery had an apparent association with normonatremia. Moreover, multivariable analyses revealed the following independent risk factors for IAH: Total urinary output over the preceding three days [odds ratio (OR) = 1.09; 95%CI: 1.02-1.17; P = 0.014], enteral nutrition (EN) sodium content of 500 mg (OR = 2.93; 95%CI: 1.13-7.60; P = 0.027), and EN sodium content of 670 mg (OR = 6.19; 95%CI: 1.75-21.98; P = 0.005) were positively correlated with the development of IAH. Notably, the area under the curve for total urinary output over the preceding three days was 0.800 (95%CI: 0.678-0.922, P = 0.001). Furthermore, maximum serum sodium levels, the duration of hypernatremia, and varying sodium correction rates were significantly associated with 28-day in-hospital mortality in septic shock patients (P < 0.05). CONCLUSION The present findings illustrate that elevated serum sodium level was significantly associated with a poor prognosis in septic shock patients in the ICU. It is highly recommended that hypernatremia be considered a potentially important prognostic indicator for the outcome of septic shock.
Collapse
Affiliation(s)
- Mai-Qing Shi
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Jun Chen
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Fu-Hai Ji
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Hao Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Ke Peng
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Jun Wang
- Intensive Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Chun-Lei Fan
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Xu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
2
|
Yakoub M, Rahman M, Kleimann P, Hoffe J, Feige M, Bouvain P, Alter C, Kluczny JI, Reidel S, Nederlof R, Hering L, Argov D, Arifaj D, Kantauskaite M, Meister J, Kleinewietfeld M, Rump LC, Jantsch J, Flögel U, Müller DN, Temme S, Stegbauer J. Transient High Salt Intake Promotes T-Cell-Mediated Hypertensive Vascular Injury. Hypertension 2024; 81:2415-2429. [PMID: 39411864 DOI: 10.1161/hypertensionaha.124.23115] [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: 04/02/2024] [Accepted: 09/18/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Dietary high salt (HS) intake has a strong impact on cardiovascular diseases. Here, we investigated the link between HS-aggravated immune responses and the development of hypertensive vascular disease. METHODS ApolipoproteinE-deficient mice were transiently treated with HS (1% NaCl) via drinking water for 2 weeks, followed by a washout period, and subsequent Ang II (angiotensin II) infusion (1000 ng/kg per min for 10 days) to induce abdominal aortic aneurysms/dissections and inflammation. RESULTS While transient HS intake alone triggered nonpathologic infiltration of activated T cells into the aorta, subsequent Ang II infusion increased mortality and the incidence of abdominal aortic aneurysms/dissections and atherosclerosis compared with hypertensive control mice. There were no differences in blood pressure between both groups. In transient HS-treated hypertensive mice, the aortic injury was associated with increased inflammation, accumulation of neutrophils, monocytes, CD69+CD4+ T cells, as well as CD4+ and CD8+ memory T cells. Mechanistically, transient HS intake increased expression levels of aortic RORγt as well as splenic CD4+TH17 and CD8+TC1 T cells in Ang II-treated mice. Isolated aortas of untreated mice were incubated with supernatants of TH17, TH1, or TC1 cells polarized in vitro under HS or normal conditions which revealed that secreted factors of HS-differentiated TH17 and TC1 cells, but not TH1 cells accelerated endothelial dysfunction. CONCLUSIONS Our data suggest that transient HS intake induces a subclinical T-cell-mediated aortic immune response, which is enhanced by Ang II. We propose a 2-hit model, in which HS acts as a predisposing factor to enhance hypertension-induced TH17 and TC1 polarization and aortic disease.
Collapse
Affiliation(s)
- Mina Yakoub
- Department of Nephrology, Faculty of Medicine, University Hospital (M.Y., M.R., J.H., M.F., L.H., D. Argov, D. Arifaj, M. Kantauskaite, L.C.R., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Masudur Rahman
- Department of Nephrology, Faculty of Medicine, University Hospital (M.Y., M.R., J.H., M.F., L.H., D. Argov, D. Arifaj, M. Kantauskaite, L.C.R., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Patricia Kleimann
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology (P.K., P.B., U.F., S.T.), Heinrich-Heine-University, Düsseldorf, Germany
- Department of Molecular Cardiology (P.K., P.B., C.A.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Jasmina Hoffe
- Department of Nephrology, Faculty of Medicine, University Hospital (M.Y., M.R., J.H., M.F., L.H., D. Argov, D. Arifaj, M. Kantauskaite, L.C.R., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Milena Feige
- Department of Nephrology, Faculty of Medicine, University Hospital (M.Y., M.R., J.H., M.F., L.H., D. Argov, D. Arifaj, M. Kantauskaite, L.C.R., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Pascal Bouvain
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology (P.K., P.B., U.F., S.T.), Heinrich-Heine-University, Düsseldorf, Germany
- Department of Molecular Cardiology (P.K., P.B., C.A.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Christina Alter
- Department of Molecular Cardiology (P.K., P.B., C.A.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Jennifer Isabel Kluczny
- Department of Anaesthesiology, Faculty of Medicine, University Hospital (J.-I.K., S.T.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Sophia Reidel
- Institut für Herz-Kreislauf-Physiologie (S.R., R.N.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Rianne Nederlof
- Institut für Herz-Kreislauf-Physiologie (S.R., R.N.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Lydia Hering
- Department of Nephrology, Faculty of Medicine, University Hospital (M.Y., M.R., J.H., M.F., L.H., D. Argov, D. Arifaj, M. Kantauskaite, L.C.R., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Doron Argov
- Department of Nephrology, Faculty of Medicine, University Hospital (M.Y., M.R., J.H., M.F., L.H., D. Argov, D. Arifaj, M. Kantauskaite, L.C.R., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Denada Arifaj
- Department of Nephrology, Faculty of Medicine, University Hospital (M.Y., M.R., J.H., M.F., L.H., D. Argov, D. Arifaj, M. Kantauskaite, L.C.R., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Marta Kantauskaite
- Department of Nephrology, Faculty of Medicine, University Hospital (M.Y., M.R., J.H., M.F., L.H., D. Argov, D. Arifaj, M. Kantauskaite, L.C.R., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Jaroslawna Meister
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Researc at Heinrich Heine University, Düsseldorf, Germany (J.M.)
- German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany (J.M.)
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC) (M. Kleinewietfeld), Hasselt University, Diepenbeek, Belgium
- Department of Immunology, Biomedical Research Institute (M. Kleinewietfeld), Hasselt University, Diepenbeek, Belgium
- University Multiple Sclerosis Center (UMSC) (M. Kleinewietfeld), Hasselt University, Diepenbeek, Belgium
| | - Lars Christian Rump
- Department of Nephrology, Faculty of Medicine, University Hospital (M.Y., M.R., J.H., M.F., L.H., D. Argov, D. Arifaj, M. Kantauskaite, L.C.R., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital Düsseldorf (L.C.R., U.F., S.T., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Jonathan Jantsch
- Institute for Medical Microbiology, Immunology and Hygiene, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany (J.J.)
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology (P.K., P.B., U.F., S.T.), Heinrich-Heine-University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital Düsseldorf (L.C.R., U.F., S.T., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Dominik N Müller
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (D.N.M.)
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany (D.N.M.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (D.N.M.)
- DZHK (German Centre for Cardiovascular Research), Germany (D.N.M.)
| | - Sebastian Temme
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology (P.K., P.B., U.F., S.T.), Heinrich-Heine-University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital Düsseldorf (L.C.R., U.F., S.T., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
- Department of Anaesthesiology, Faculty of Medicine, University Hospital (J.-I.K., S.T.), Heinrich-Heine-University, Düsseldorf, Germany
| | - Johannes Stegbauer
- Department of Nephrology, Faculty of Medicine, University Hospital (M.Y., M.R., J.H., M.F., L.H., D. Argov, D. Arifaj, M. Kantauskaite, L.C.R., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital Düsseldorf (L.C.R., U.F., S.T., J.S.), Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
3
|
Yang C, Wang T, Zhao C, Lu J, Shen R, Li G, Zhao J. Causal relationship of salt intake with osteoarthritis: A Mendelian randomization analysis. Medicine (Baltimore) 2024; 103:e40497. [PMID: 39560570 PMCID: PMC11575978 DOI: 10.1097/md.0000000000040497] [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: 09/11/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
Recent studies have demonstrated a correlation between salt intake (SI) and various diseases. However, it remains uncertain whether the relationship between SI (including salt added to food and sodium levels in urine) and benign osteoarthritis is causal. To investigate this, we conducted a 2-sample Mendelian randomization (MR) analysis to estimate the causal impact of SI on osteoarthritis (OA). A genome-wide association study of salt added to food and sodium in urine was used as the exposure, while hip osteoarthritis, knee osteoarthritis, and rheumatoid arthritis were defined as the outcomes. Inverse variance weighting (IVW) was used to calculate causal estimates, and sensitivity analyses were performed using methods including weighted mode, weighted median, MR-Egger, and Bayesian weighted MR. All statistical analyses were conducted using R software. Our results, primarily based on the IVW method, support the existence of a causal relationship between salt added to food and knee osteoarthritis (KOA). Specifically, salt added to food was associated with a decreased risk of KOA (OR = 1.248, P = .024, 95% CI: 1.030-1.512). This study is the first MR investigation exploring the causal relationship between salt added to food and KOA, potentially providing new insights and a theoretical basis for the prevention and treatment of KOA in the future.
Collapse
Affiliation(s)
- Chengrui Yang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Tieqiang Wang
- Cangzhou Hospital of Integrated Chinese and Western Medicine, Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, Cangzhou, Hebei, China
| | - Chunzhi Zhao
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jiawei Lu
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Runbin Shen
- Cangzhou Hospital of Integrated Chinese and Western Medicine, Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, Cangzhou, Hebei, China
| | - Guoliang Li
- Cangzhou Hospital of Integrated Chinese and Western Medicine, Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, Cangzhou, Hebei, China
| | - Jianyong Zhao
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- Cangzhou Hospital of Integrated Chinese and Western Medicine, Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, Cangzhou, Hebei, China
| |
Collapse
|
4
|
Vulin M, Muller A, Drenjančević I, Šušnjara P, Mihaljević Z, Stupin A. High dietary salt intake attenuates nitric oxide mediated endothelium-dependent vasodilation and increases oxidative stress in pregnancy. J Hypertens 2024; 42:672-684. [PMID: 38230612 DOI: 10.1097/hjh.0000000000003645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
OBJECTIVE This study aimed to investigate the impact of dietary salt intake during normal pregnancy on maternal microvascular and macrovascular endothelium-dependent reactivity and oxidative stress level. MATERIALS AND METHODS In this cross-sectional study, based on their 24-h urinary sodium excretion, pregnant women (37-40 weeks of gestation) were divided into three groups: normal salt (<5.75 g/day, N = 12), high salt (5.75-10.25 g/day, N = 36), and very high salt (VHS;>10.25 g/day, N = 17). Forearm skin microvascular reactivity in response to vascular occlusion, local heating (LTH) and iontophoresis of acetylcholine (AChID), as well as brachial artery flow mediated dilation (FMD) were measured. Serum nitric oxide, endocan, 8-iso-prostaglandin F2α (8-iso-PGF2α), thiobarbituric acid reactive substances (TBARS), and ferric-reducing ability of plasma assay were measured as biomarkers of endothelial function/activation and oxidative stress. RESULTS Brachial artery FMD, microvascular AChID, and LTH were significantly decreased in VHS compared with NS group, while LTH was also decreased in normal salt compared with high salt group. Nitric oxide was significantly decreased in both high salt and VHS groups compared with normal salt. Endocan, 8-iso-PGF2α, and TBARS were significantly increased in VHS compared with the normal salt group. CONCLUSION High dietary salt intake is associated with decreased nitric oxide mediated endothelium-dependent vasodilation in peripheral microcirculation and macrocirculation of healthy pregnant women due to increased oxidative stress.
Collapse
Affiliation(s)
- Martina Vulin
- Department of Gynaecology and Obstetrics, University Hospital Centre Osijek
- Department of Gynaecology and Obstetrics, Faculty of Medicine Osijek
| | - Andrijana Muller
- Department of Gynaecology and Obstetrics, University Hospital Centre Osijek
- Department of Gynaecology and Obstetrics, Faculty of Medicine Osijek
| | - Ines Drenjančević
- Department of Physiology and Immunology, Faculty of Medicine Osijek
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Petar Šušnjara
- Department of Physiology and Immunology, Faculty of Medicine Osijek
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Zrinka Mihaljević
- Department of Physiology and Immunology, Faculty of Medicine Osijek
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Ana Stupin
- Department of Physiology and Immunology, Faculty of Medicine Osijek
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| |
Collapse
|
5
|
Miyauchi H, Geisberger S, Luft FC, Wilck N, Stegbauer J, Wiig H, Dechend R, Jantsch J, Kleinewietfeld M, Kempa S, Müller DN. Sodium as an Important Regulator of Immunometabolism. Hypertension 2024; 81:426-435. [PMID: 37675565 PMCID: PMC10863658 DOI: 10.1161/hypertensionaha.123.19489] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Salt sensitivity concerns blood pressure alterations after a change in salt intake (sodium chloride). The heart is a pump, and vessels are tubes; sodium can affect both. A high salt intake increases cardiac output, promotes vascular dysfunction and capillary rarefaction, and chronically leads to increased systemic vascular resistance. More recent findings suggest that sodium also acts as an important second messenger regulating energy metabolism and cellular functions. Besides endothelial cells and fibroblasts, sodium also affects innate and adaptive immunometabolism, immune cell function, and influences certain microbes and microbiota-derived metabolites. We propose the idea that the definition of salt sensitivity should be expanded beyond high blood pressure to cellular and molecular salt sensitivity.
Collapse
Affiliation(s)
- Hidetaka Miyauchi
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.M., S.G., F.C.L., N.W., R.D., S.K., D.N.M.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Germany (H.M., F.C.L., N.W., R.D., D.N.M.)
- Experimental and Clinical Research Center, a joint cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (H.M., F.C.L., N.W., R.D., D.N.M.)
- German Centre for Cardiovascular Research, Partner Site Berlin, Germany (H.M., N.W., R.D., D.N.M.)
| | - Sabrina Geisberger
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.M., S.G., F.C.L., N.W., R.D., S.K., D.N.M.)
| | - Friedrich C. Luft
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.M., S.G., F.C.L., N.W., R.D., S.K., D.N.M.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Germany (H.M., F.C.L., N.W., R.D., D.N.M.)
- Experimental and Clinical Research Center, a joint cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (H.M., F.C.L., N.W., R.D., D.N.M.)
| | - Nicola Wilck
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.M., S.G., F.C.L., N.W., R.D., S.K., D.N.M.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Germany (H.M., F.C.L., N.W., R.D., D.N.M.)
- Experimental and Clinical Research Center, a joint cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (H.M., F.C.L., N.W., R.D., D.N.M.)
- German Centre for Cardiovascular Research, Partner Site Berlin, Germany (H.M., N.W., R.D., D.N.M.)
| | - Johannes Stegbauer
- Department of Nephrology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, Düsseldorf, Germany (J.S.)
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany (J.S.)
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Norway (H.W.)
| | - Ralf Dechend
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.M., S.G., F.C.L., N.W., R.D., S.K., D.N.M.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Germany (H.M., F.C.L., N.W., R.D., D.N.M.)
- Experimental and Clinical Research Center, a joint cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (H.M., F.C.L., N.W., R.D., D.N.M.)
- German Centre for Cardiovascular Research, Partner Site Berlin, Germany (H.M., N.W., R.D., D.N.M.)
- HELIOS Clinic, Department of Cardiology and Nephrology, Berlin, Germany (R.D.)
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, Germany (J.J.)
- Institute for Medical Microbiology, Immunology, and Hygiene, and Center for Molecular Medicine Cologne, University Hospital Cologne and Faculty of Medicine, University of Cologne, Germany (J.J.)
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, Hasselt University, Diepenbeek, Belgium (M.K.)
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium (M.K.)
- University Multiple Sclerosis Center, Hasselt University/Campus Diepenbeek, Belgium (M.K.)
| | - Stefan Kempa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.M., S.G., F.C.L., N.W., R.D., S.K., D.N.M.)
| | - Dominik N. Müller
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.M., S.G., F.C.L., N.W., R.D., S.K., D.N.M.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Germany (H.M., F.C.L., N.W., R.D., D.N.M.)
- Experimental and Clinical Research Center, a joint cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (H.M., F.C.L., N.W., R.D., D.N.M.)
- German Centre for Cardiovascular Research, Partner Site Berlin, Germany (H.M., N.W., R.D., D.N.M.)
| |
Collapse
|
6
|
Lin T, Jiang D, Chen W, Lin JS, Zhang X, Chen C, Hsu C, Lai L, Chen P, Yang K, Sansing LH, Chang C. Trained immunity induced by high-salt diet impedes stroke recovery. EMBO Rep 2023; 24:e57164. [PMID: 37965920 PMCID: PMC10702837 DOI: 10.15252/embr.202357164] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 10/22/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023] Open
Abstract
A high-salt diet (HSD) elicits sustained sterile inflammation and worsens tissue injury. However, how this occurs after stroke, a leading cause of morbidity and mortality, remains unknown. Here, we report that HSD impairs long-term brain recovery after intracerebral hemorrhage, a severe form of stroke, despite salt withdrawal prior to the injury. Mechanistically, HSD induces innate immune priming and training in hematopoietic stem and progenitor cells (HSPCs) by downregulation of NR4a family and mitochondrial oxidative phosphorylation. This training compromises alternative activation of monocyte-derived macrophages (MDMs) without altering the initial inflammatory responses of the stroke brain. Healthy mice transplanted with bone marrow from HSD-fed mice retain signatures of reduced MDM reparative functions, further confirming a persistent form of innate immune memory that originates in the bone marrow. Loss of NR4a1 in macrophages recapitulates HSD-induced negative impacts on stroke outcomes while gain of NR4a1 enables stroke recovery in HSD animals. Together, we provide the first evidence that links HSD-induced innate immune memory to the acquisition of persistent dysregulated inflammatory responses and unveils NR4a1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Tze‐Yen Lin
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Danye Jiang
- Department of NeurologyMcGovern Medical School at the University of Texas Health Science Center in HoustonHoustonTXUSA
| | - Wan‐Ru Chen
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
- School of MedicineNational Taiwan University College of MedicineTaipeiTaiwan
| | - Jhih Syuan Lin
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Xin‐Yu Zhang
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Chih‐Hung Chen
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Chia‐Lang Hsu
- Department of Medical ResearchNational Taiwan University HospitalTaipeiTaiwan
| | - Liang‐Chuan Lai
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Ping‐Hung Chen
- Department and Graduate Institute of Biochemistry and Molecular BiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Kai‐Chien Yang
- Department and Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Lauren H Sansing
- Department of NeurologyYale University School of MedicineNew HavenCTUSA
| | - Che‐Feng Chang
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| |
Collapse
|
7
|
Oppelaar JJ, Bouwmeester TA, Silova AA, Collard D, Wouda RD, van Duin RE, Rorije NMG, Olde Engberink RHG, Danser AHJ, van den Born BJH, Vogt L. Salt-sensitive trait of normotensive individuals is associated with altered autonomous cardiac regulation: a randomized controlled intervention study. Am J Physiol Renal Physiol 2023; 325:F707-F716. [PMID: 37795535 DOI: 10.1152/ajprenal.00076.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/07/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023] Open
Abstract
Blood pressure (BP) responses to sodium intake show great variation, discriminating salt-sensitive (SS) from salt-resistant (SR) individuals. The pathophysiology behind salt sensitivity is still not fully elucidated. We aimed to investigate salt-induced effects on body fluid, vascular tone, and autonomic cardiac response with regard to BP change in healthy normotensive individuals. We performed a randomized crossover study in 51 normotensive individuals with normal body mass index and estimated glomerular filtration rate. Subjects followed both a low-Na+ diet (LSD, <50 mmol/day) and a high-Na+ diet (HSD, >200 mmol/day). Cardiac output, systemic vascular resistance (SVR), and cardiac autonomous activity, through heart rate variability and cross-correlation baroreflex sensitivity (xBRS), were assessed with noninvasive continuous finger BP measurements. In a subset, extracellular volume (ECV) was assessed by iohexol measurements. Subjects were characterized as SS if mean arterial pressure (MAP) increased ≥3 mmHg after HSD. After HSD, SS subjects (25%) showed a 6.1-mmHg (SD 1.9) increase in MAP. No differences between SS and SR in body weight, cardiac output, or ECV were found. SVR was positively correlated with Delta BP (r = 0.31, P = 0.03). xBRS and heart rate variability were significantly higher in SS participants compared to SR participants after both HSD and LSD. Sodium loading did not alter heart rate variability within groups. Salt sensitivity in normotensive individuals is associated with an inability to decrease SVR upon high salt intake that is accompanied by alterations in autonomous cardiac regulation, as reflected by decreased xBRS and heart rate variability. No discriminatory changes upon high salt were observed among salt-sensitive individuals in body weight and ECV.NEW & NOTEWORTHY Extracellular fluid expansion in normotensive individuals after salt loading is present in both salt-sensitive and salt-resistant individuals and is not discriminatory to the blood pressure response to sodium loading in a steady-state measurement. In normotensive subjects, the ability to sufficiently vasodilate seems to play a pivotal role in salt sensitivity. In a normotensive cohort, differences in sympathovagal balance are also present in low-salt conditions rather than being affected by salt loading. Whereas treatment and prevention of salt-sensitive blood pressure increase are mostly focused on renal sodium handling and extracellular volume regulation, our study suggests that an inability to adequately vasodilate and altered autonomous cardiac functioning are additional key players in the pathophysiology of salt-sensitive blood pressure increase.
Collapse
Affiliation(s)
- Jetta J Oppelaar
- Section of Nephrology, Department of Internal Medicine, Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Microcirculation, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Thomas A Bouwmeester
- Section of Vascular Medicine, Department of Internal Medicine, Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Anastasia A Silova
- Section of Nephrology, Department of Internal Medicine, Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
| | - Didier Collard
- Section of Vascular Medicine, Department of Internal Medicine, Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Rosa D Wouda
- Section of Nephrology, Department of Internal Medicine, Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Microcirculation, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Robert E van Duin
- Section of Nephrology, Department of Internal Medicine, Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Microcirculation, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Nienke M G Rorije
- Section of Nephrology, Department of Internal Medicine, Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Microcirculation, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Rik H G Olde Engberink
- Section of Nephrology, Department of Internal Medicine, Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Microcirculation, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bert-Jan H van den Born
- Section of Vascular Medicine, Department of Internal Medicine, Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Liffert Vogt
- Section of Nephrology, Department of Internal Medicine, Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Microcirculation, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Vogt L, Marques FZ, Fujita T, Hoorn EJ, Danser AHJ. Novel mechanisms of salt-sensitive hypertension. Kidney Int 2023; 104:690-697. [PMID: 37454911 DOI: 10.1016/j.kint.2023.06.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
A high dietary sodium-consumption level is considered the most important lifestyle factor that can be modified to help prevent an increase in blood pressure and the development of hypertension. Despite numerous studies over the past decades, the pathophysiology explaining why some people show a salt-sensitive blood pressure response and others do not is incompletely understood. Here, a brief overview of the latest mechanistic insights is provided, focusing on the mononuclear phagocytic system and inflammation, the gut-kidney axis, and epigenetics. The article also discusses the effects of 3 types of novel drugs on salt-sensitive hypertension-sodium-glucose cotransporter 2 inhibitors, nonsteroidal mineralocorticoid receptor antagonists, and aldosterone synthase inhibitors. The conclusion is that besides kidney-centered mechanisms, vasoconstrictor mechanisms are also relevant for both the understanding and treatment of this blood pressure phenotype.
Collapse
Affiliation(s)
- Liffert Vogt
- Department of Internal Medicine, Section of Nephrology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, and Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Toshiro Fujita
- Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
9
|
Wang T, Masedunskas A, Willett WC, Fontana L. Vegetarian and vegan diets: benefits and drawbacks. Eur Heart J 2023; 44:3423-3439. [PMID: 37450568 PMCID: PMC10516628 DOI: 10.1093/eurheartj/ehad436] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023] Open
Abstract
Plant-based diets have become increasingly popular thanks to their purported health benefits and more recently for their positive environmental impact. Prospective studies suggest that consuming vegetarian diets is associated with a reduced risk of developing cardiovascular disease (CVD), diabetes, hypertension, dementia, and cancer. Data from randomized clinical trials have confirmed a protective effect of vegetarian diets for the prevention of diabetes and reductions in weight, blood pressure, glycosylated haemoglobin and low-density lipoprotein cholesterol, but to date, no data are available for cardiovascular event rates and cognitive impairment, and there are very limited data for cancer. Moreover, not all plant-based foods are equally healthy. Unhealthy vegetarian diets poor in specific nutrients (vitamin B12, iron, zinc, and calcium) and/or rich in highly processed and refined foods increase morbidity and mortality. Further mechanistic studies are desirable to understand whether the advantages of healthy, minimally processed vegetarian diets represent an all-or-nothing phenomenon and whether consuming primarily plant-based diets containing small quantities of animal products (e.g. pesco-vegetarian or Mediterranean diets) has beneficial, detrimental, or neutral effects on cardiometabolic health outcomes. Further, mechanistic studies are warranted to enhance our understanding about healthy plant-based food patterns and the biological mechanisms linking dietary factors, CVD, and other metabolic diseases.
Collapse
Affiliation(s)
- Tian Wang
- Charles Perkins Center, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Andrius Masedunskas
- Charles Perkins Center, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Walter C Willett
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Luigi Fontana
- Charles Perkins Center, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Department of Clinical and Experimental Sciences, Brescia University, Brescia, Lombardy, Italy
| |
Collapse
|
10
|
Copur S, Peltek IB, Mutlu A, Tanriover C, Kanbay M. A new immune disease: systemic hypertension. Clin Kidney J 2023; 16:1403-1419. [PMID: 37664577 PMCID: PMC10469084 DOI: 10.1093/ckj/sfad059] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Indexed: 09/05/2023] Open
Abstract
Systemic hypertension is the most common medical comorbidity affecting the adult population globally, with multiple associated outcomes including cerebrovascular diseases, cardiovascular diseases, vascular calcification, chronic kidney disease, metabolic syndrome and mortality. Despite advancements in the therapeutic field approximately one in every five adult patients with hypertension is classified as having treatment-resistant hypertension, indicating the need for studies to provide better understanding of the underlying pathophysiology and the need for more therapeutic targets. Recent pre-clinical studies have demonstrated the role of the innate and adaptive immune system including various cell types and cytokines in the pathophysiology of hypertension. Moreover, pre-clinical studies have indicated the potential beneficial effects of immunosuppressant medications in the control of hypertension. Nevertheless, it is unclear whether such pathophysiological mechanisms and therapeutic alternatives are applicable to human subjects, while this area of research is undoubtedly a rapidly growing field.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ibrahim B Peltek
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ali Mutlu
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Cem Tanriover
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Mehmet Kanbay
- Department of Medicine, Section of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| |
Collapse
|
11
|
Mongelos MA, Sosa FN, Pineda GE, Fiorentino G, Santiago A, Abelleyro MM, Rossetti LC, Exeni R, De Brasi CD, Palermo MS, Ramos MV. Assessment of interleukin-10 promoter variant (-1082A/G) and cytokine production in patients with hemolytic uremic syndrome. Front Pediatr 2023; 11:1210158. [PMID: 37425258 PMCID: PMC10327435 DOI: 10.3389/fped.2023.1210158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Hemolytic uremic syndrome (HUS) is a condition that results in acute kidney failure mainly in children, which is caused by Shiga toxin-producing Escherichia coli and inflammatory response. Although anti-inflammatory mechanisms are triggered, studies on the implication in HUS are scarce. Interleukin-10 (IL-10) regulates inflammation in vivo, and the interindividual differences in its expression are related to genetic variants. Notably, the single nucleotide polymorphism (SNP) rs1800896 -1082 (A/G), located in the IL-10 promoter, regulates cytokine expression. Methods Plasma and peripheral blood mononuclear cells (PBMC) were collected from healthy children and HUS patients exhibiting hemolytic anemia, thrombocytopenia, and kidney damage. Monocytes identified as CD14+ cells were analyzed within PBMC by flow cytometry. IL-10 levels were quantified by ELISA, and SNP -1082 (A/G) was analyzed by allele-specific PCR. Results Circulating IL-10 levels were increased in HUS patients, but PBMC from these patients exhibited a lower capacity to secrete this cytokine compared with those from healthy children. Interestingly, there was a negative association between the circulating levels of IL-10 and inflammatory cytokine IL-8. We observed that circulating IL-10 levels were threefold higher in HUS patients with -1082G allele in comparison to AA genotype. Moreover, there was relative enrichment of GG/AG genotypes in HUS patients with severe kidney failure. Discussion Our results suggest a possible contribution of SNP -1082 (A/G) to the severity of kidney failure in HUS patients that should be further evaluated in a larger cohort.
Collapse
Affiliation(s)
- Micaela Aldana Mongelos
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental (CONICET)—Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Fernando Nicolás Sosa
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental (CONICET)—Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Gonzalo Ezequiel Pineda
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental (CONICET)—Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Gabriela Fiorentino
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental (CONICET)—Academia Nacional de Medicina, Buenos Aires, Argentina
- Departamento de Nefrología, Diálisis y Trasplante, Hospital del Niño Prof. Dr. Ramón Exeni, San Justo, Argentina
| | - Adriana Santiago
- Departamento de Nefrología, Diálisis y Trasplante, Hospital del Niño Prof. Dr. Ramón Exeni, San Justo, Argentina
| | - Miguel Martín Abelleyro
- Laboratorio de Genética Molecular de la Hemofilia, Instituto de Medicina Experimental (CONICET)—Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Liliana Carmen Rossetti
- Laboratorio de Genética Molecular de la Hemofilia, Instituto de Medicina Experimental (CONICET)—Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Ramón Exeni
- Departamento de Nefrología, Diálisis y Trasplante, Hospital del Niño Prof. Dr. Ramón Exeni, San Justo, Argentina
| | - Carlos Daniel De Brasi
- Laboratorio de Genética Molecular de la Hemofilia, Instituto de Medicina Experimental (CONICET)—Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Marina Sandra Palermo
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental (CONICET)—Academia Nacional de Medicina, Buenos Aires, Argentina
| | - María Victoria Ramos
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental (CONICET)—Academia Nacional de Medicina, Buenos Aires, Argentina
| |
Collapse
|
12
|
Ilesanmi-Oyelere BL, Kruger MC. Associations between dietary patterns and an array of inflammation biomarkers and plasma lipid profile in postmenopausal women. BMC Womens Health 2023; 23:256. [PMID: 37173732 PMCID: PMC10176721 DOI: 10.1186/s12905-023-02417-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 05/06/2023] [Indexed: 05/15/2023] Open
Abstract
OBJECTIVE AND DESIGN In this cross-sectional study, evaluation of the association between four dietary patterns, nutrients and food intakes and an array of systemic inflammation biomarkers and lipid profile among 80 New Zealand postmenopausal women were conducted. MATERIALS Eighty postmenopausal women participated in the study. A validated food frequency questionnaire was used to collect nutrients and food intake. Four dietary patterns were identified by principal component analysis (PCA) and plasma samples collected for inflammatory biomarkers and lipid profile measures. RESULTS There were negative correlations between intake of dietary fibre, soluble and insoluble non-starch polysaccharides (NSP), vitamin C and niacin and with almost all the inflammatory markers for the whole group. Vegetables, tea/coffee and especially fruit intake were negatively correlated with the inflammatory biomarkers in the whole group. A high intake of Pattern 1 (potato, bread, and fruit pattern) was associated with a low risk of high interferon (IFN)-α2, IFN-λ, interleukin (IL)-6 and IL-8 levels while a high intake of Pattern 3 (fast-food pattern) was associated high risk of IFN-α2 levels. Multiple linear regression showed a negative correlation between Pattern 2 (soups and vegetables pattern) and levels of C-reactive protein (CRP) as well as ferritin. A positive association was observed between Pattern 3 (fast-food pattern) and CRP levels. Positive correlation was also observed between Pattern 2 and high-density lipoprotein (HDL) and total cholesterol (TC) levels, Pattern 4 (meat and vegetables pattern) was however negatively correlated with TC, low-density lipoprotein (LDL) and TC/HDL ratio. CONCLUSIONS The result of this study reinforces the contribution and role of diet in modifying inflammation in postmenopausal women.
Collapse
Affiliation(s)
- Bolaji L Ilesanmi-Oyelere
- School of Health Sciences, College of Health, Massey University, Palmerston North, New Zealand.
- Human Nutrition and Dietetics Department, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates.
| | - Marlena C Kruger
- School of Health Sciences, College of Health, Massey University, Palmerston North, New Zealand
| |
Collapse
|
13
|
Linder BA, Babcock MC, Pollin KU, Watso JC, Robinson AT. Short-term high-salt consumption does not influence resting or exercising heart rate variability but increases MCP-1 concentration in healthy young adults. Am J Physiol Regul Integr Comp Physiol 2023; 324:R666-R676. [PMID: 36939211 PMCID: PMC10110701 DOI: 10.1152/ajpregu.00240.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/21/2023]
Abstract
High salt consumption increases blood pressure (BP) and cardiovascular disease risk by altering autonomic function and increasing inflammation. However, it is unclear whether salt manipulation alters resting and exercising heart rate variability (HRV), a noninvasive measure of autonomic function, in healthy young adults. The purpose of this investigation was to determine whether short-term high-salt intake 1) alters HRV at rest, during exercise, or exercise recovery and 2) increases the circulating concentration of the inflammatory biomarker monocyte chemoattractant protein 1 (MCP-1). With the use of a randomized, placebo-controlled, crossover study, 20 participants (8 females; 24 ± 4 yr old, 110 ± 10/64 ± 8 mmHg) consumed salt (3,900 mg sodium) or placebo capsules for 10 days each separated by ≥2 wk. We assessed HRV during 10 min of baseline rest, 50 min of cycling (60% V̇o2peak), and recovery. We quantified HRV using the standard deviation of normal-to-normal RR intervals, the root mean square of successive differences (RMSSD), and additional time and frequency domain metrics of HRV. Plasma samples were collected to assess MCP-1 concentration. No main effect of high salt or condition × time interaction was observed for HRV metrics. However, acute exercise reduced HRV (e.g., RMSSD time: P < 0.001, condition: P = 0.877, interaction: P = 0.422). High salt elevated plasma MCP-1 (72.4 ± 12.5 vs. 78.14 ± 14.7 pg/mL; P = 0.010). Irrespective of condition, MCP-1 was moderately associated (P values < 0.05) with systolic (r = 0.32) and mean BP (r = 0.33). Short-term high-salt consumption does not affect HRV; however, it increases circulating MCP-1, which may influence BP in young adults.
Collapse
Affiliation(s)
- Braxton A Linder
- Neurovascular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Matthew C Babcock
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, United States
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kamila U Pollin
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, United States
- War Related Illness and Injury Study Center, Washington DC Veteran Affairs Medical Center, Washington, District of Columbia, United States
| | - Joseph C Watso
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, United States
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, United States
| | - Austin T Robinson
- Neurovascular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama, United States
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, United States
| |
Collapse
|
14
|
Zhang X, Liang Y, Jiang J, Lu C, Shi F, Cao Q, Zhang Y, Diao H. A High-Salt Diet Exacerbates Liver Fibrosis through Enterococcus-Dependent Macrophage Activation. Microbiol Spectr 2023; 11:e0340322. [PMID: 36786636 PMCID: PMC10100947 DOI: 10.1128/spectrum.03403-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/14/2023] [Indexed: 02/15/2023] Open
Abstract
People consume more salt than the recommended levels due to poor dietary practices. The effects of long-term consumption of high-salt diets (HSD) on liver fibrosis are unclear. This study aimed to explore the impact of HSD on liver fibrosis. In this study, a carbon tetrachloride (CCL4)-induced liver fibrosis mouse model was used to evaluate fibrotic changes in the livers of mice fed a normal diet (ND) and an HSD. The HSD exacerbated liver injury and fibrosis. Moreover, the protein expression levels of transforming growth factor β (TGF-β), tumor necrosis factor alpha (TNF-α), and monocyte chemoattractant protein 1 (MCP-1) were significantly higher in the HSD group than in the normal group. The proportion of macrophages and activation significantly increased in the livers of HSD-fed mice. Meanwhile, the number of macrophages significantly increased in the small intestinal lamina propria of HSD-fed mice. The levels of profibrotic factors also increased in the small intestine of HSD-fed mice. Additionally, HSD increased the profibrotic chemokines and monocyte chemoattractant levels in the portal vein blood. Further characterization suggested that the HSD decreased the expression of tight junction proteins (ZO-1 and CLDN1), enhancing the translocation of bacteria. Enterococcus promoted liver injury and inflammation. In vitro experiments demonstrated that Enterococcus induced macrophage activation through the NF-κB pathway, thus promoting the expression of fibrosis-related genes, leading to liver fibrogenesis. Similarly, Enterococcus disrupted the gut microbiome in vivo and significantly increased the fibrotic markers, TGF-β, and alpha smooth muscle actin (α-SMA) expression in the liver. IMPORTANCE This study further confirms that Enterococcus induce liver fibrosis in mice. These results indicate that an HSD can exacerbate liver fibrosis by altering the gut microbiota composition, thus impairing intestinal barrier function. Therefore, this may serve as a new target for liver fibrosis therapy and gut microbiota management.
Collapse
Affiliation(s)
- Xujun Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yan Liang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingjing Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Chong Lu
- Department of Gastroenterology, Jinhua Hospital of Zhejiang University, Jinhua, Zhejiang, China
| | - Fan Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Qingyi Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanhui Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Cao Y, Fan Y, Li F, Hao Y, Kong Y, Chen C, Hao X, Han D, Li G, Wang Z, Song C, Han J, Zeng H. Phenotypic and functional alterations of monocyte subsets with aging. Immun Ageing 2022; 19:63. [PMID: 36514074 PMCID: PMC9745938 DOI: 10.1186/s12979-022-00321-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND It has been widely accepted that monocytes are one of the central mediators contributing to inflammaging. However, it remains unclear whether aged monocytes, similar to aged T cells, have characteristics of hyperactivation and increased expression of co-inhibitory molecules. METHODS Peripheral blood mononuclear cells (PBMCs) were isolated from young (21-40 years old), middle-aged (41-60 years old), and older human subjects (> 60 years old). Flow cytometry was used to monitor changes in the expression of surface molecules of monocyte subsets and cytokine-producing capacity. RESULTS We observed increased tumor necrosis factor-α: TNF-α and decreased interleukin-6 (IL-6) production in monocytes from older adults compared with young and middle-aged adults. Older adults had a greater percentage of intermediate and non-classical monocyte subsets, along with increased levels of the immune activation markers human leukocyte antigen-DR (HLA-DR), and adhesion molecules cluster of differentiation molecule 11b (CD11b) and L-selectin (CD62L). Furthermore, we observed increased C-C motif chemokine receptor 2 (CCR2) expression on classical monocytes and decreased C-X3-C motif chemokine receptor 1 (CX3CR1) expression on non-classical monocytes in older adult subjects. The expression of co-inhibitory receptors was reduced on monocyte subsets in older adults. CONCLUSIONS Circulating monocytes in older adults exhibit increased expression of activation, adhesion, and migration markers, but decreased expression of co-inhibitory molecules.
Collapse
Affiliation(s)
- Yu Cao
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China ,grid.508381.70000 0004 0647 272XBeijing Institute of Infectious Diseases, Beijing, 100015 China ,grid.24696.3f0000 0004 0369 153XNational Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China
| | - Yang Fan
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China ,grid.508381.70000 0004 0647 272XBeijing Institute of Infectious Diseases, Beijing, 100015 China ,grid.24696.3f0000 0004 0369 153XNational Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China
| | - Fangyuan Li
- grid.414367.3Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China ,grid.414367.3Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China
| | - Yu Hao
- grid.414367.3Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China ,grid.414367.3Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China
| | - Yaxian Kong
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China ,grid.508381.70000 0004 0647 272XBeijing Institute of Infectious Diseases, Beijing, 100015 China ,grid.24696.3f0000 0004 0369 153XNational Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China
| | - Chen Chen
- grid.414367.3Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China ,grid.414367.3Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China
| | - Xing Hao
- grid.411606.40000 0004 1761 5917Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029 China
| | - Dannuo Han
- grid.411606.40000 0004 1761 5917Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029 China
| | - Guoli Li
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China ,grid.508381.70000 0004 0647 272XBeijing Institute of Infectious Diseases, Beijing, 100015 China ,grid.24696.3f0000 0004 0369 153XNational Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China
| | - Zengtao Wang
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China ,grid.508381.70000 0004 0647 272XBeijing Institute of Infectious Diseases, Beijing, 100015 China ,grid.24696.3f0000 0004 0369 153XNational Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China
| | - Chuan Song
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China ,grid.508381.70000 0004 0647 272XBeijing Institute of Infectious Diseases, Beijing, 100015 China ,grid.24696.3f0000 0004 0369 153XNational Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China
| | - Junyan Han
- grid.414367.3Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China ,grid.414367.3Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China
| | - Hui Zeng
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China ,grid.508381.70000 0004 0647 272XBeijing Institute of Infectious Diseases, Beijing, 100015 China ,grid.24696.3f0000 0004 0369 153XNational Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China ,grid.414367.3Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China ,grid.414367.3Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China
| |
Collapse
|
16
|
Basalely AM, Griffin R, Gist KM, Guillet R, Askenazi DJ, Charlton JR, Selewski DT, Fuloria M, Kaskel FJ, Reidy KJ. Association of early dysnatremia with mortality in the neonatal intensive care unit: results from the AWAKEN study. J Perinatol 2022; 42:1353-1360. [PMID: 34775486 PMCID: PMC10228559 DOI: 10.1038/s41372-021-01260-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/12/2021] [Accepted: 10/20/2021] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To determine the association of dysnatremia in the first postnatal week and risk of acute kidney injury (AKI) and mortality. STUDY DESIGN A secondary analysis of 1979 neonates in the AWAKEN cohort evaluated the association of dysnatremia with (1) AKI in the first postnatal week and (2) mortality, utilizing time-varying Cox proportional hazard models. RESULT Dysnatremia developed in 50.2% of the cohort and was not associated with AKI. Mortality was associated with hyponatremia (HR 2.15, 95% CI 1.07-4.31), hypernatremia (HR 4.23, 95% CI 2.07-8.65), and combined hypo/hypernatremia (HR 6.39, 95% CI 2.01-14.01). In stratified models by AKI-status, hypernatremia and hypo/hypernatremia increased risk of mortality in neonates without AKI. CONCLUSION Dysnatremia within the first postnatal week was associated with increased risk of mortality. Hypernatremia and combined hypo/hypernatremia remained significantly associated with mortality in neonates without AKI. This may reflect fluid strategies kidney injury independent of creatinine and urine-output defined AKI, and/or systemic inflammation.
Collapse
Affiliation(s)
- Abby M Basalely
- Division of Pediatric Nephrology, Department of Pediatrics, Cohen Children's Medical Center of New York, New Hyde Park, NY, USA.
- Division of Nephrology, Department of Pediatrics, The Children's Hospital at Montefiore/Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Russell Griffin
- Pediatric and Infant Center for Acute Nephrology (PICAN) Department of Pediatrics, University of Alabama, Birmingham, AL, USA
| | - Katja M Gist
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ronnie Guillet
- Division of Neonatology, University of Rochester Medical Center, Rochester, NY, USA
| | - David J Askenazi
- Pediatric and Infant Center for Acute Nephrology (PICAN) Department of Pediatrics, University of Alabama, Birmingham, AL, USA
| | - Jennifer R Charlton
- Division of Nephrology, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - David T Selewski
- Division of Nephrology, Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Mamta Fuloria
- Division of Pediatric Neonatology, Department of Pediatrics, The Children's Hospital at Montefiore/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Frederick J Kaskel
- Division of Nephrology, Department of Pediatrics, The Children's Hospital at Montefiore/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kimberly J Reidy
- Division of Nephrology, Department of Pediatrics, The Children's Hospital at Montefiore/Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
17
|
Association of Hypernatremia with Immune Profiles and Clinical Outcomes in Adult Intensive Care Unit Patients with Sepsis. Biomedicines 2022; 10:biomedicines10092285. [PMID: 36140385 PMCID: PMC9496274 DOI: 10.3390/biomedicines10092285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/05/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Both hypernatremia and an abnormal immune response may increase hospital mortality in patients with sepsis. This study examined the association of hypernatremia with abnormal immune response and mortality in 520 adult patients with sepsis in an intensive care unit (ICU). We compared the mortality and ex vivo lipopolysaccharide (LPS)-induced inflammatory response differences among patients with hyponatremia, eunatremia, and hypernatremia, as well as between patients with acquired hypernatremia on ICU day 3 and those with sustained eunatremia over first three ICU days. Compared with eunatremia or hyponatremia, hypernatremia led to higher 7 day, 14 day, 28 day, and hospital mortality rates (p = 0.030, 0.009, 0.010, and 0.033, respectively). Compared with sustained eunatremia, acquired hypernatremia led to higher 7, 14, and 28 day mortality rates (p = 0.019, 0.042, and 0.028, respectively). The acquired hypernatremia group nonsignificantly trended toward increased hospital mortality (p = 0.056). Day 1 granulocyte colony-stimulating factor (G-CSF) and tumor necrosis factor (TNF) α levels were relatively low in patients with hypernatremia (p = 0.020 and 0.010, respectively) but relatively high in patients with acquired hypernatremia (p = 0.049 and 0.009, respectively). Thus, in ICU-admitted septic patients, hypernatremia on admission and in ICU-acquired hypernatremia were both associated with higher mortality. The higher mortality in patients with hypernatremia on admission was possibly related to the downregulation of G-CSF and TNF-α secretion after endotoxin stimulation. Compared to sustained eunatremia, acquired hypernatremia showed immunoparalysis at first and then hyperinflammation on day 3.
Collapse
|
18
|
Li X, Alu A, Wei Y, Wei X, Luo M. The modulatory effect of high salt on immune cells and related diseases. Cell Prolif 2022; 55:e13250. [PMID: 35747936 PMCID: PMC9436908 DOI: 10.1111/cpr.13250] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The adverse effect of excessive salt intake has been recognized in decades. Researchers have mainly focused on the association between salt intake and hypertension. However, studies in recent years have proposed the existence of extra-renal sodium storage and provided insight into the immunomodulatory function of sodium. OBJECTIVES In this review, we discuss the modulatory effects of high salt on various innate and adaptive immune cells and immune-regulated diseases. METHODS We identified papers through electronic searches of PubMed database from inception to March 2022. RESULTS An increasing body of evidence has demonstrated that high salt can modulate the differentiation, activation and function of multiple immune cells. Furthermore, a high-salt diet can increase tissue sodium concentrations and influence the immune responses in microenvironments, thereby affecting the development of immune-regulated diseases, including hypertension, multiple sclerosis, cancer and infections. These findings provide a novel mechanism for the pathology of certain diseases and indicate that salt might serve as a target or potential therapeutic agent in different disease contexts. CONCLUSION High salt has a profound impact on the differentiation, activation and function of multiple immune cells. Additionally, an HSD can modulate the development of various immune-regulated diseases.
Collapse
Affiliation(s)
- Xian Li
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Aqu Alu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Min Luo
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Wenstedt EFE, Peters Sengers H, Boekholdt SM, Khaw K, Wareham NJ, van den Born BH, Vogt L. Relationship of Sodium Intake With Granulocytes, Renal and Cardiovascular Outcomes in the Prospective EPIC-Norfolk Cohort. J Am Heart Assoc 2022; 11:e023727. [PMID: 35730648 PMCID: PMC9333397 DOI: 10.1161/jaha.121.023727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Experimental studies show that high-sodium intake affects the innate immune system, among others with increased circulating granulocytes. Whether this relationship exists on a population level and whether this relates to disease outcomes is unclear. We aimed to test the hypotheses that (1) sodium intake is associated with granulocytes on a population level; (2) granulocytes are associated with the presence of hypertension and both cardiovascular and renal outcomes; and (3) the relation between high-sodium intake and these outcomes is mediated by granulocytes. Methods and Results We performed an analysis in 13 804 participants from the prospective EPIC (European Prospective Investigation into Cancer)-Norfolk cohort, with a mean age of 58 years and median follow-up of 19.3 years. Analyses were carried out using calculated estimated sodium intake and sodium-to-potassium ratios from spot urines at baseline. The main outcomes were hypertension at baseline, and composite cardiovascular (mortality or cardiovascular events) and renal (mortality or renal events) outcomes during follow-up. Sodium intake and urine sodium-to-potassium ratio were positively associated with circulating granulocyte concentrations after adjustment for confounders (β=0.03; P=0.028 and β=0.06; P<0.001, respectively). Granulocytes significantly mediated the associations of, respectively, sodium intake and urine sodium-to-potassium ratio with hypertension at baseline, and cardiovascular and renal outcomes. Conclusions Sodium intake is positively associated with circulating granulocyte concentrations, and higher granulocyte concentrations associate with worse long-term cardiovascular and renal outcomes. Given the recently established immune-modulating effects of sodium and the role of immune cells in both cardiovascular and renal disease, causality for this pathway may need consideration in further studies.
Collapse
Affiliation(s)
- Eliane F. E. Wenstedt
- Department of Internal MedicineSection of NephrologyAmsterdam UMCUniversity of AmsterdamAmsterdam Cardiovascular SciencesAmsterdamThe Netherlands
| | - Hessel Peters Sengers
- Amsterdam UMCUniversity of AmsterdamCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands,Amsterdam UMCThe Amsterdam Institute for Infection and ImmunityAmsterdamThe Netherlands
| | | | - Kay‐Tee Khaw
- Department of Public Health and Primary CareUniversity of CambridgeUnited Kingdom,MRC Epidemiology UnitCambridgeUnited Kingdom
| | | | - Bert‐Jan H. van den Born
- Department of Internal MedicineSection of Vascular MedicineAmsterdam UMCUniversity of AmsterdamAmsterdam Cardiovascular SciencesAmsterdamThe Netherlands
| | - Liffert Vogt
- Department of Internal MedicineSection of NephrologyAmsterdam UMCUniversity of AmsterdamAmsterdam Cardiovascular SciencesAmsterdamThe Netherlands
| |
Collapse
|
20
|
Attaye I, Warmbrunn MV, Boot ANAF, van der Wolk SC, Hutten BA, Daams JG, Herrema H, Nieuwdorp M. A Systematic Review and Meta-analysis of Dietary Interventions Modulating Gut Microbiota and Cardiometabolic Diseases-Striving for New Standards in Microbiome Studies. Gastroenterology 2022; 162:1911-1932. [PMID: 35151697 DOI: 10.1053/j.gastro.2022.02.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND & AIMS Cardiometabolic diseases (CMDs) have shared properties and causes. Insulin resistance is a risk factor and characteristic of CMDs and has been suggested to be modulated by plasma metabolites derived from gut microbiota (GM). Because diet is among the most important modulators of GM, we performed a systematic review of the literature to assess whether CMDs can be modulated via dietary interventions targeting the GM. METHODS A systematic review of the literature for clinical studies was performed on Ovid MEDLINE and Ovid Embase. Studies were assessed for risk of bias and patterns of intervention effects. A meta-analysis with random effects models was used to evaluate the effect of dietary interventions on clinical outcomes. RESULTS Our search yielded 4444 unique articles, from which 15 randomized controlled trials and 6 nonrandomized clinical trials were included. The overall risk of bias was high in all studies. In general, most dietary interventions changed the GM composition, but no consistent effect could be found. Results of the meta-analyses showed that only diastolic blood pressure is decreased across interventions compared with controls (mean difference: -3.63 mm Hg; 95% confidence interval, -7.09 to -0.17; I2 = 0%, P = .04) and that a high-fiber diet was associated with reduced triglyceride levels (mean difference: -0.69 mmol/L; 95% confidence interval, -1.36 to -0.02; I2 = 59%, P = .04). Other CMD parameters were not affected. CONCLUSIONS Dietary interventions modulate GM composition, blood pressure, and circulating triglycerides. However, current studies have a high methodological heterogeneity and risk of bias. Well-designed and controlled studies are thus necessary to better understand the complex interaction between diet, microbiome, and CMDs. PROSPERO CRD42020188405.
Collapse
Affiliation(s)
- Ilias Attaye
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Moritz V Warmbrunn
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Aureline N A F Boot
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Suze C van der Wolk
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Barbara A Hutten
- Department of Epidemiology and Data Science, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Joost G Daams
- Medical Library, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Hilde Herrema
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands.
| |
Collapse
|
21
|
Linder BA, Mehrer JD, Bunsawat K. Salt skintitle "salty skin: Where excess sodium goes for a rendezvous". J Physiol 2022; 600:3025-3027. [PMID: 35621318 DOI: 10.1113/jp283274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Braxton A Linder
- Neurovascular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, AL, 36849, USA
| | - Justin D Mehrer
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, 19711, USA
| | - Kanokwan Bunsawat
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, 84132, USA.,Geriatric Research, Education, and Clinical Center, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, 84148, USA
| |
Collapse
|
22
|
Schmitz J, Brauns N, Hüsing AM, Flechsig M, Glomb T, Bräsen JH, Haller H, von Vietinghoff S. Renal medullary osmolytes NaCl and urea differentially modulate human tubular cell cytokine expression and monocyte recruitment. Eur J Immunol 2022; 52:1258-1272. [DOI: 10.1002/eji.202149723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/20/2022] [Accepted: 05/06/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Jessica Schmitz
- Nephropathology Unit Institute for Pathology University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Nicolas Brauns
- Department of Internal Medicine Division of Nephrology and Hypertension University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Anne M. Hüsing
- Department of Internal Medicine Division of Nephrology and Hypertension University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Martina Flechsig
- Department of Internal Medicine Division of Nephrology and Hypertension University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Thorsten Glomb
- Core Facility Transcriptomics Hannover Medical School Hannover Germany
| | - Jan Hinrich Bräsen
- Nephropathology Unit Institute for Pathology University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Hermann Haller
- Department of Internal Medicine Division of Nephrology and Hypertension University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Sibylle von Vietinghoff
- Department of Internal Medicine Division of Nephrology and Hypertension University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
- Nephrology Section First Medical Clinic University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| |
Collapse
|
23
|
Barnett AM, Babcock MC, Watso JC, Migdal KU, Gutiérrez OM, Farquhar WB, Robinson AT. High dietary salt intake increases urinary NGAL excretion and creatinine clearance in healthy young adults. Am J Physiol Renal Physiol 2022; 322:F392-F402. [PMID: 35157527 PMCID: PMC8934673 DOI: 10.1152/ajprenal.00240.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 02/02/2022] [Accepted: 02/11/2022] [Indexed: 11/22/2022] Open
Abstract
In rodents and older patients with elevated blood pressure (BP), high dietary sodium increases excretion of biomarkers of kidney injury, but it is unclear whether this effect occurs in healthy young adults. The purpose of this study was to determine whether short-term high dietary salt increases urinary excretion of the kidney injury biomarkers neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1) in healthy young adults. Twenty participants participated in a double-blind, placebo-controlled, randomized crossover study. For 10 days each, participants were asked to consume salt (3,900 mg sodium) or placebo capsules. We measured BP during each visit, obtained 24-h urine samples for measurements of electrolytes, NGAL, and KIM-1, and assessed creatinine clearance. Compared with placebo, salt loading increased daily urinary sodium excretion (placebo: 130.3 ± 62.4 mmol/24 h vs. salt: 287.2 ± 72.0 mmol/24 h, P < 0.01). There was no difference in mean arterial BP (placebo: 77 ± 7 mmHg vs. salt: 77 ± 6 mmHg, P = 0.83) between conditions. However, salt loading increased the urinary NGAL excretion rate (placebo: 59.8 ± 44.4 ng/min vs. salt: 80.8 ± 49.5 ng/min, P < 0.01) and increased creatinine clearance (placebo: 110.5 ± 32.9 mL/min vs. salt: 145.0 ± 24.9 mL/min, P < 0.01). Urinary KIM-1 excretion was not different between conditions. In conclusion, in healthy young adults 10 days of dietary salt loading increased creatinine clearance and increased urinary excretion of the kidney injury biomarker marker NGAL but not KIM-1.NEW & NOTEWORTHY In healthy young adults, 10 days of dietary salt loading increased creatinine clearance and increased urinary excretion of the kidney injury biomarker marker neutrophil gelatinase-associated lipocalin despite no change in resting blood pressure.
Collapse
Affiliation(s)
- Alex M Barnett
- Neurovascular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama
| | - Matthew C Babcock
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
- Division of Geriatric Medicine, University of Colorado-Anschutz Medical Campus, Aurora, Colorado
| | - Joseph C Watso
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kamila U Migdal
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
- War Related Illness and Injury Study Center, Washington DC Department of Veteran Affairs Medical Center, Washington, District of Columbia
| | - Orlando M Gutiérrez
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - William B Farquhar
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
| | - Austin T Robinson
- Neurovascular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama
| |
Collapse
|
24
|
Ha YJ, Ji E, Lee JH, Kim JH, Park EH, Chung SW, Chang SH, Yoo JJ, Kang EH, Ahn S, Song YW, Lee YJ. High Estimated 24-Hour Urinary Sodium Excretion Is Related to Symptomatic Knee Osteoarthritis: A Nationwide Cross-Sectional Population-Based Study. J Nutr Health Aging 2022; 26:581-589. [PMID: 35718867 DOI: 10.1007/s12603-022-1804-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES High salt intake results in various harmful effects on human health including hypertension, cardiovascular disease, and reduced bone density. Despite this, there are very few studies in the literature that have investigated the association between sodium intake and osteoarthritis (OA). Therefore, we aimed to explore these associations in a Korean population. METHODS This study used cross-sectional data from adult subjects aged 50-75 years from two consecutive periods of the Korean National Health and Nutrition Examination Survey V-VII (2010-2011 and 2014-2016). The estimated 24-hour urinary sodium excretion (24HUNa) was used as a surrogate marker of salt intake. In the 2010-2011 dataset, knee OA (KOA) was defined as the presence of the radiographic features of OA and knee pain. The association between KOA and salt intake was analysed using univariable and multivariable logistic regression methods. For the sensitivity analysis, the same procedures were conducted on subjects with self-reported OA (SR-OA) with knee pain in the 2010-2011 dataset and any site SR-OA in the 2014-2016 dataset. RESULTS Subjects with KOA had significantly lower energy intake, but higher 24HUNa than those without KOA. The restricted cubic spline plots demonstrated a J-shaped distribution between 24HUNa and prevalent KOA. When 24HUNa was stratified into five groups (<2, 2-3, 3-4, 4-5 and ≥5 g/day), subjects with high sodium intake (≥5 g/day) had a higher risk of KOA (odds ratio [OR] = 1.64, 95% confidence interval [CI] 1.03-2.62) compared to the reference group (3-4 g/day) after adjusting for covariates. The sensitivity analysis based on SR-OA with knee pain showed that high sodium intake was also significantly associated with increased prevalence of OA (OR = 1.84, 95% CI 1.10-3.10) compared with the reference group. Regarding SR-OA at any site in the 2014-2016 dataset, estimated 24HUNa showed a significantly positive association with the presence of SR-OA after adjusting for potential confounders. CONCLUSIONS This nationwide Korean representative study showed a significant association between symptomatic KOA and high sodium intake (≥5 g/day). Avoidance of a diet high in salt might be beneficial as a non-pharmacologic therapy for OA.
Collapse
Affiliation(s)
- Y-J Ha
- Yun Jong Lee, M.D., Ph.D., Department of Internal Medicine, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beongil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea, Tel.: +82-31-787-7049, Fax.: +82-31-787-4051, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Sahinoz M, Elijovich F, Ertuglu LA, Ishimwe J, Pitzer A, Saleem M, Mwesigwa N, Kleyman TR, Laffer CL, Kirabo A. Salt Sensitivity of Blood Pressure in Blacks and Women: A Role of Inflammation, Oxidative Stress, and Epithelial Na + Channel. Antioxid Redox Signal 2021; 35:1477-1493. [PMID: 34569287 PMCID: PMC8713266 DOI: 10.1089/ars.2021.0212] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/11/2022]
Abstract
Significance: Salt sensitivity of blood pressure (SSBP) is an independent risk factor for mortality and morbidity due to cardiovascular disease, and disproportionately affects blacks and women. Several mechanisms have been proposed, including exaggerated activation of sodium transporters in the kidney leading to salt retention and water. Recent Advances: Recent studies have found that in addition to the renal epithelium, myeloid immune cells can sense sodium via the epithelial Na+ channel (ENaC), which leads to activation of the nicotinamide adenine dinucleotide phosphate oxidase enzyme complex, increased fatty acid oxidation, and production of isolevuglandins (IsoLGs). IsoLGs are immunogenic and contribute to salt-induced hypertension. In addition, aldosterone-mediated activation of ENaC has been attributed to the increased SSBP in women. The goal of this review is to highlight mechanisms contributing to SSBP in blacks and women, including, but not limited to increased activation of ENaC, fatty acid oxidation, and inflammation. Critical Issues: A critical barrier to progress in management of SSBP is that its diagnosis is not feasible in the clinic and is limited to expensive and laborious research protocols, which makes it difficult to investigate. Yet without understanding the underlying mechanisms, this important risk factor remains without treatment. Future Directions: Further studies are needed to understand the mechanisms that contribute to differential blood pressure responses to dietary salt and find feasible diagnostic tools. This is extremely important and may go a long way in mitigating the racial and sex disparities in cardiovascular outcomes. Antioxid. Redox Signal. 35, 1477-1493.
Collapse
Affiliation(s)
- Melis Sahinoz
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Fernando Elijovich
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lale A. Ertuglu
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeanne Ishimwe
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ashley Pitzer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mohammad Saleem
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Naome Mwesigwa
- Department of Medicine and Dentistry, Kampala International University, Kampala, Uganda
| | - Thomas R. Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cheryl L. Laffer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
26
|
Fei L, Ren X, Yu H, Zhan Y. Targeting the CCL2/CCR2 Axis in Cancer Immunotherapy: One Stone, Three Birds? Front Immunol 2021; 12:771210. [PMID: 34804061 PMCID: PMC8596464 DOI: 10.3389/fimmu.2021.771210] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/19/2021] [Indexed: 12/15/2022] Open
Abstract
CCR2 is predominantly expressed by monocytes/macrophages with strong proinflammatory functions, prompting the development of CCR2 antagonists to dampen unwanted immune responses in inflammatory and autoimmune diseases. Paradoxically, CCR2-expressing monocytes/macrophages, particularly in tumor microenvironments, can be strongly immunosuppressive. Thus, targeting the recruitment of immunosuppressive monocytes/macrophages to tumors by CCR2 antagonism has recently been investigated as a strategy to modify the tumor microenvironment and enhance anti-tumor immunity. We present here that beneficial effects of CCR2 antagonism in the tumor setting extend beyond blocking chemotaxis of suppressive myeloid cells. Signaling within the CCL2/CCR2 axis shows underappreciated effects on myeloid cell survival and function polarization. Apart from myeloid cells, T cells are also known to express CCR2. Nevertheless, tissue homing of Treg cells among T cell populations is preferentially affected by CCR2 deficiency. Further, CCR2 signaling also directly enhances Treg functional potency. Thus, although Tregs are not the sole type of T cells expressing CCR2, the net outcome of CCR2 antagonism in T cells favors the anti-tumor arm of immune responses. Finally, the CCL2/CCR2 axis directly contributes to survival/growth and invasion/metastasis of many types of tumors bearing CCR2. Together, CCR2 links to two main types of suppressive immune cells by multiple mechanisms. Such a CCR2-assoicated immunosuppressive network is further entangled with paracrine and autocrine CCR2 signaling of tumor cells. Strategies to target CCL2/CCR2 axis as cancer therapy in the view of three types of CCR2-expessing cells in tumor microenvironment are discussed.
Collapse
Affiliation(s)
- Liyang Fei
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Xiaochen Ren
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Haijia Yu
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Yifan Zhan
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| |
Collapse
|
27
|
Frieler RA, Vigil TM, Song J, Leung C, Lumeng CN, Mortensen RM. High-fat and high-sodium diet induces metabolic dysfunction in the absence of obesity. Obesity (Silver Spring) 2021; 29:1868-1881. [PMID: 34549547 PMCID: PMC8571049 DOI: 10.1002/oby.23264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Excess dietary fat and sodium (NaCl) are both associated with obesity and metabolic dysfunction. In mice, high NaCl has been shown to block high-fat (HF) diet-induced weight gain. Here, the impact of an HF/NaCl diet on metabolic function in the absence of obesity was investigated. METHODS Wild-type mice were administered chow, NaCl (4%), HF, and HF/NaCl diets. Metabolic analysis was performed by measuring fasted blood glucose and insulin levels and by glucose tolerance test and insulin tolerance test. RESULTS After 10 weeks on diets, male and female mice on the HF diet gained weight, and HF/NaCl mice had significantly reduced weight gain similar to chow-fed mice. In the absence of obesity, HF/NaCl mice had significantly elevated fasting blood glucose and impaired glucose control during glucose tolerance tests. Both NaCl and HF/NaCl mice had decreased pancreas and β-cell mass. Administration of NaCl in drinking water did not protect mice from HF-diet-induced weight gain and obesity. Further analysis revealed that longer administration of HF/NaCl diets for 20 weeks resulted in significant weight gain and insulin resistance. CONCLUSIONS The data demonstrate that despite early inhibitory effects on fat deposition and weight gain, an HF/NaCl diet does not prevent the metabolic consequences of HF diet consumption.
Collapse
Affiliation(s)
- Ryan A. Frieler
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| | - Thomas M. Vigil
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| | - Jianrui Song
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| | - Christy Leung
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| | - Carey N. Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI
| | - Richard M. Mortensen
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW It is accepted that several chronic diseases are associated with inflammation. Dietary habits and the consumption of certain nutrients have been shown to influence inflammation, reflected by inflammatory cytokines. In this narrative review, we discuss currently developed tools to assess the inflammatory potential of diets and compare them with established tools. RECENT FINDINGS Four new indices were recently developed. The Inflammatory Score of the Diet is a modified version of the established Dietary Inflammatory Index. The novel Empirical Dietary Inflammatory Index works without previous dietary intake assessment and the Anti-Inflammatory Diet Index was specifically developed in a northern European population. The Dietary and Lifestyle Inflammation Scores addresses additional confounders. The informative value of dietary indices relies on the accuracy and completeness of dietary intake assessment. SUMMARY Dietary inflammatory indices are important tools to assess, compare and validate the inflammatory potential of diets across populations without the need for biomarker assessments. They allow to investigate associations between an (anti)-inflammatory diet with disease risk and course. Although the Dietary Inflammatory Index remains the most used index worldwide, currently developed indices allow more flexibility, have a different focus or simplify assessment. Additional foods, that were recently shown to modulate inflammation, but are not (fully) considered yet, may deserve more attention in the future.
Collapse
Affiliation(s)
- Lina Samira Bahr
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, ECRC Experimental and Clinical Research Center
| | - Kristina Franz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Research Group on Geriatrics
| | - Anja Mähler
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, ECRC Experimental and Clinical Research Center
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
29
|
Basdeki ED, Kollias A, Mitrou P, Tsirimiagkou C, Georgakis MK, Chatzigeorgiou A, Argyris A, Karatzi K, Manios Y, Sfikakis PP, Protogerou AD. Does Sodium Intake Induce Systemic Inflammatory Response? A Systematic Review and Meta-Analysis of Randomized Studies in Humans. Nutrients 2021; 13:2632. [PMID: 34444792 PMCID: PMC8399701 DOI: 10.3390/nu13082632] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/13/2021] [Accepted: 07/27/2021] [Indexed: 12/29/2022] Open
Abstract
Experimental studies suggest that sodium induced inflammation might be another missing link leading to atherosclerosis. To test the hypothesis that high daily sodium intake induces systemic inflammatory response in humans, we performed a systematic review according to PRISMA guidelines of randomized controlled trials (RCTs) that examined the effect of high versus low sodium dose (HSD vs. LSD), as defined per study, on plasma circulating inflammatory biomarkers. Eight RCTs that examined CRP, TNF-a and IL-6 were found. Meta-analysis testing the change of each biomarker in HSD versus LSD was possible for CRP (n = 5 studies), TNF-a (n = 4 studies) and IL-6 (n = 4 studies). The pooled difference (95% confidence intervals) per biomarker was for: CRP values of 0.1(-0.3, 0.4) mg/L; TNF-a -0.7(-5.0, 3.6) pg/mL; IL-6 -1.1(-3.3 to 1.1) pg/mL. Importantly, there was inconsistency between RCTs regarding major population characteristics and the applied methodology, including a very wide range of LSD (460 to 6740 mg/day) and HSD (2800 to 7452 mg/day). Although our results suggest that the different levels of daily sodium intake are not associated with significant changes in the level of systemic inflammation in humans, this outcome may result from methodological issues. Based on these identified methodological issues we propose that future RCTs should focus on young healthy participants to avoid confounding effects of comorbidities, should have three instead of two arms (very low, "normal" and high) of daily sodium intake with more than 100 participants per arm, whereas an intervention duration of 14 days is adequate.
Collapse
Affiliation(s)
- Eirini D. Basdeki
- Cardiovascular Prevention & Research Unit, Clinic & Laboratory of Pathophysiology, Department of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.D.B.); (C.T.); (A.A.)
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17671 Kallithea, Greece;
| | - Anastasios Kollias
- Hypertension Center STRIDE-7, Third Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, 11527 Athens, Greece;
| | | | - Christiana Tsirimiagkou
- Cardiovascular Prevention & Research Unit, Clinic & Laboratory of Pathophysiology, Department of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.D.B.); (C.T.); (A.A.)
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17671 Kallithea, Greece;
| | - Marios K. Georgakis
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilians University, 81377 Munich, Germany;
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National & Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Antonios Argyris
- Cardiovascular Prevention & Research Unit, Clinic & Laboratory of Pathophysiology, Department of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.D.B.); (C.T.); (A.A.)
| | - Kalliopi Karatzi
- Department of Food Science and Human Nutrition, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece;
| | - Yannis Manios
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17671 Kallithea, Greece;
| | - Petros P. Sfikakis
- First Department of Propaedeutic and Internal Medicine, Athens University Medical School, Laiko Hospital, 11527 Athens, Greece;
| | - Athanase D. Protogerou
- Cardiovascular Prevention & Research Unit, Clinic & Laboratory of Pathophysiology, Department of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.D.B.); (C.T.); (A.A.)
| |
Collapse
|
30
|
Sun H, Hu L, Fan Z. β2 integrin activation and signal transduction in leukocyte recruitment. Am J Physiol Cell Physiol 2021; 321:C308-C316. [PMID: 34133240 DOI: 10.1152/ajpcell.00560.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Leukocyte recruitment is a critical step in the pathogenesis of inflammatory and immunological responses. Cell adhesion molecules (CAMs) are involved in controlling cell movements and the recruitment process, and the integrin family of CAMs plays a key role. During cell movement, integrin function is dynamically and precisely regulated. However, this balance might be broken under pathological conditions. Thus, the functional regulation and molecular mechanisms of integrins related to diseases are often a focus of research. Integrin β2 is one of the most commonly expressed integrins in leukocytes that mediate leukocyte adhesion and migration, and it plays an important role in immune responses and inflammation. In this review, we focus on specific functions of integrin β2 in leukocyte recruitment, the conformational changes and signal transduction of integrin β2 activation, the similarities between murine and human factors, and how new insights into these processes can inform future therapies for inflammation and immune diseases.
Collapse
Affiliation(s)
- Hao Sun
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Liang Hu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut
| |
Collapse
|
31
|
Jobin K, Müller DN, Jantsch J, Kurts C. Sodium and its manifold impact on our immune system. Trends Immunol 2021; 42:469-479. [PMID: 33962888 DOI: 10.1016/j.it.2021.04.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 12/19/2022]
Abstract
The Western diet is rich in salt, and a high salt diet (HSD) is suspected to be a risk factor for cardiovascular diseases. It is now widely accepted that an experimental HSD can stimulate components of the immune system, potentially exacerbating certain autoimmune diseases, or alternatively, improving defenses against certain infections, such as cutaneous leishmaniasis. However, recent findings show that an experimental HSD may also aggravate other infections (e.g., pyelonephritis or systemic listeriosis). Here, we discuss the modulatory effects of a HSD on the microbiota, metabolic signaling, hormonal responses, local sodium concentrations, and their effects on various immune cell types in different tissues. We describe how these factors are integrated, resulting either in immune stimulation or suppression in various tissues and disease settings.
Collapse
Affiliation(s)
- Katarzyna Jobin
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany; Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, Würzburg, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, and Max Delbruck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg and University of Regensburg, Regensburg, Germany.
| | - Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany; Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia.
| |
Collapse
|
32
|
Wenstedt EFE, van Croonenburg TJ, van den Born BJH, Van den Bossche J, Hooijmans CR, Vogt L. The effect of macrophage-targeted interventions on blood pressure - a systematic review and meta-analysis of preclinical studies. Transl Res 2021; 230:123-138. [PMID: 33166696 DOI: 10.1016/j.trsl.2020.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/08/2020] [Accepted: 11/03/2020] [Indexed: 01/01/2023]
Abstract
An increasing body of evidence shows a role for macrophages and monocytes (as their precursors) in hypertension, but with conflicting results with regard to whether they are protective or harmful. Therefore, we systematically reviewed the effect of macrophage interventions on blood pressure in animal models, to explore which factors determine the blood pressure increasing vs. decreasing effect. A search in PubMED and EMBASE yielded 9620 records, 26 of which were included. Eighteen studies (involving 22 different experiments (k = 22)) performed macrophage depletion, whereas 12 studies specifically deleted certain macrophage proteins. The blood pressure effects of macrophage depletion were highly various and directed toward both directions, as expected, which could not be reduced to differences in animal species or methods of hypertension induction. Prespecified subgroup analysis did reveal a potential role for the route in which the macrophage-depleting agent is being administrated (intraperitoneal vs intravenous subgroup difference of P = 0.07 (k = 22), or P < 0.001 in studies achieving considerable (ie, >50%) depletion (k = 18)). Along with findings from specific macrophage protein deletion studies-showing that deletion of one single macrophage protein (like TonEBP, endothelin-B, EP4, NOX-2 and the angiotensin II type 1 receptor) can alter blood pressure responses to hypertensive stimuli-the indication that each route has its specific depletion pattern regarding targeted tissues and macrophage phenotypes suggests a determinative role for these features. These hypothesis-generating results encourage more detailed depletion characterization of each technique by direct experimental comparisons, providing a chance to obtain more knowledge on which macrophages are beneficial versus detrimental in hypertension development.
Collapse
Affiliation(s)
- Eliane F E Wenstedt
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Thirza J van Croonenburg
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Bert-Jan H van den Born
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jan Van den Bossche
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Carlijn R Hooijmans
- Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE), Department of Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Liffert Vogt
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Jung R, Wild J, Ringen J, Karbach S, Wenzel P. Innate Immune Mechanisms of Arterial Hypertension and Autoimmune Disease. Am J Hypertens 2021; 34:143-153. [PMID: 32930786 DOI: 10.1093/ajh/hpaa145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/15/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022] Open
Abstract
The immune system is indispensable in the development of vascular dysfunction and hypertension. The interplay between immune cells and the vasculature, kidneys, heart, and blood pressure regulating nuclei in the central nervous system results in a complex and closely interwoven relationship of the immune system with arterial hypertension. A better understanding of this interplay is necessary for optimized and individualized antihypertensive therapy. Our review article focuses on innate cells in hypertension and to what extent they impact on development and preservation of elevated blood pressure. Moreover, we address the association of hypertension with chronic autoimmune diseases. The latter are ideally suited to learn about immune-mediated mechanisms in cardiovascular disease leading to high blood pressure.
Collapse
Affiliation(s)
- Rebecca Jung
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Johannes Wild
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Julia Ringen
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Susanne Karbach
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Germany
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Germany
| |
Collapse
|
34
|
McCarthy CG, Saha P, Golonka RM, Wenceslau CF, Joe B, Vijay-Kumar M. Innate Immune Cells and Hypertension: Neutrophils and Neutrophil Extracellular Traps (NETs). Compr Physiol 2021; 11:1575-1589. [PMID: 33577121 DOI: 10.1002/cphy.c200020] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Uncontrolled immune system activation amplifies end-organ injury in hypertension. Nonetheless, the exact mechanisms initiating this exacerbated inflammatory response, thereby contributing to further increases in blood pressure (BP), are still being revealed. While participation of lymphoid-derived immune cells has been well described in the hypertension literature, the mechanisms by which myeloid-derived innate immune cells contribute to T cell activation, and subsequent BP elevation, remains an active area of investigation. In this article, we critically analyze the literature to understand how monocytes, macrophages, dendritic cells, and polymorphonuclear leukocytes, including mast cells, eosinophils, basophils, and neutrophils, contribute to hypertension and hypertension-associated end-organ injury. The most abundant leukocytes, neutrophils, are indisputably increased in hypertension. However, it is unknown how (and why) they switch from critical first responders of the innate immune system, and homeostatic regulators of BP, to tissue-damaging, pro-hypertensive mediators. We propose that myeloperoxidase-derived pro-oxidants, neutrophil elastase, neutrophil extracellular traps (NETs), and interactions with other innate and adaptive immune cells are novel mechanisms that could contribute to the inflammatory cascade in hypertension. We further posit that the gut microbiota serves as a set point for neutropoiesis and their function. Finally, given that hypertension appears to be a key risk factor for morbidity and mortality in COVID-19 patients, we put forth evidence that neutrophils and NETs cause cardiovascular injury post-coronavirus infection, and thus may be proposed as an intriguing therapeutic target for high-risk individuals. © 2021 American Physiological Society. Compr Physiol 11:1575-1589, 2021.
Collapse
Affiliation(s)
- Cameron G McCarthy
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Piu Saha
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Rachel M Golonka
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Camilla F Wenceslau
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Bina Joe
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Matam Vijay-Kumar
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| |
Collapse
|
35
|
Wenstedt EFE, Remmerswaal EBM, van der Bom-Baylon ND, Schrooten EM, Bemelman FJ, Vogt L. The effect of high-salt diet on t-lymphocyte subpopulations in healthy males-A pilot study. J Clin Hypertens (Greenwich) 2020; 22:2152-2155. [PMID: 32960505 PMCID: PMC7756710 DOI: 10.1111/jch.14049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/13/2020] [Accepted: 08/31/2020] [Indexed: 01/06/2023]
Abstract
Animal studies show that high‐salt diet affects T‐cell subpopulations, but evidence in humans is scarce and contradictory. This pilot study investigated the effect of a 2‐week high‐salt diet on T‐cell subpopulations (ie, γδ T cells, Th17 cells, and regulatory T cells) in five healthy males. The mean (SD) age of the participants was 33 (2) years, with normal body mass index, kidney function, and baseline blood pressure. In terms of phenotype, there was an isolated increase of CD69 expression in Vδ1 T cells (P = .04), which is an early activation marker. There were no statistically significant changes or trends in any of the other tested markers or in the Th17 or regulatory T‐cell subsets. The increase in CD69 was strongly correlated to increases in 24‐hour urinary sodium excretion (r = .93, P = .02). These results of this pilot may motivate the use of longer dietary salt interventions in future studies on salt and adaptive immune cells.
Collapse
Affiliation(s)
- Eliane F E Wenstedt
- Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ester B M Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nelly D van der Bom-Baylon
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Esmee M Schrooten
- Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Frederike J Bemelman
- Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Liffert Vogt
- Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|