1
|
Koubar SH, Garcia-Rivera A, Mohamed MMB, Hall JE, Hall ME, Hassanein M. Underlying Mechanisms and Treatment of Hypertension in Glomerular Diseases. Curr Hypertens Rep 2024; 26:119-130. [PMID: 37982994 DOI: 10.1007/s11906-023-01287-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2023] [Indexed: 11/21/2023]
Abstract
PURPOSE OF REVIEW This review aims to explore the underlying mechanisms that lead to hypertension in glomerular diseases and the advancements in treatment strategies and to provide clinicians with valuable insights into the pathophysiological mechanisms and evidence-based therapeutic approaches for managing hypertension in patients with glomerular diseases. RECENT FINDINGS In recent years, there have been remarkable advancements in our understanding of the immune and non-immune mechanisms that are involved in the pathogenesis of hypertension in glomerular diseases. Furthermore, this review will encompass the latest data on management strategies, including RAAS inhibition, endothelin receptor blockers, SGLT2 inhibitors, and immune-based therapies. Hypertension (HTN) and cardiovascular diseases are leading causes of mortality in glomerular diseases. The latter are intricately related with hypertension and share common pathophysiological mechanisms. Hypertension in glomerular disease represents a complex and multifaceted interplay between kidney dysfunction, immune-mediated, and non-immune-mediated pathology. Understanding the complex mechanisms involved in this relationship has evolved significantly over the years, shedding light on the pathophysiological processes underlying the development and progression of glomerular disease-associated HTN, and is crucial for developing effective therapeutic strategies and improving patients' outcomes.
Collapse
Affiliation(s)
- Sahar H Koubar
- Division of Nephrology and Hypertension, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Alejandro Garcia-Rivera
- Department of Nephrology. Hospital General Regional 46, Instituto Mexicano del Seguro Social, Guadalajara, Mexico
| | - Muner M B Mohamed
- Department of Nephrology, Ochsner Health System, New Orleans, LA, USA
- Ochsner Clinical School, The University of Queensland, Brisbane, QLD, Australia
| | - John E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael E Hall
- Division of Cardiovascular Disease, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Mohamed Hassanein
- Division of Nephrology and Hypertension, Department of Medicine, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS, USA.
| |
Collapse
|
2
|
Akbari A, Hadizadeh A, Islampanah M, Salavati Nik E, Atkin SL, Sahebkar A. COVID-19, G protein-coupled receptor, and renin-angiotensin system autoantibodies: Systematic review and meta-analysis. Autoimmun Rev 2023; 22:103402. [PMID: 37490975 DOI: 10.1016/j.autrev.2023.103402] [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: 07/11/2023] [Accepted: 07/20/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION There are an increasing number of reports of autoantibodies (AAbs) against host proteins such as G-protein coupled receptors (GPCRs) and the renin-angiotensin system (RAS) in COVID-19 disease. Here we have undertaken a systematic review and meta-analysis of all reports of AAbs against GPCRs and RAS in COVID-19 patients including those with long-COVID or post-COVID symptoms. METHODS PubMed, Embase, Web of Science, and Scopus databases were searched to find papers on the role of GPCR and RAS AAbs in the presence and severity of COVID-19 or post- COVID symptoms available through March 21, 2023. Data on the prevalence of AngII or ACE, comparing AngII or ACE between COVID-19 and non-COVID-19, or comparing AngII or ACE between COVID-19 patients with different disease stages were pooled and a meta-analysed using random- or fixed-effects models were undertaken. RESULTS The search yielded a total of 1042 articles, of which 68 studies were included in this systematic review and nine in the meta-analysis. Among 18 studies that investigated GPCRs and COVID-19 severity, 18 distinct AAbs were detected. In addition, nine AAbs were found in case reports that assessed post- COVID, and 19 AAbs were found in other studies that assessed post- COVID or long- COVID symptoms. Meta-analysis revealed a significantly higher number of seropositive ACE2 AAbs in COVID-19 patients (odds ratio = 7.766 [2.056, 29.208], p = 0.002) and particularly in severe disease (odds ratio = 11.49 [1.04, 126.86], p = 0.046), whereas AngII-AAbs seropositivity was no different between COVID-19 and control subjects (odds ratio = 2.890 [0.546-15.283], p = 0.21). CONCLUSIONS GPCR and RAS AAbs may play an important role in COVID-19 severity, the development of disease progression, long-term symptoms COVID and post- COVID symptoms.
Collapse
Affiliation(s)
- Abolfazl Akbari
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Hadizadeh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Muhammad Islampanah
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ensie Salavati Nik
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Stephen L Atkin
- Royal College of Surgeons in Ireland, Bahrain, Adliya, PO Box 15503, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Abstract
BACKGROUND Atrial fibrillation (AF) is by far the most common cardiac arrhythmia. In about 3% of individuals, AF develops as a primary disorder without any identifiable trigger (idiopathic or historically termed lone AF). In line with the emerging field of autoantibody-related cardiac arrhythmias, the objective of this study was to explore whether autoantibodies targeting cardiac ion channels can underlie unexplained AF. METHODS Peptide microarray was used to screen patient samples for autoantibodies. We compared patients with unexplained AF (n=37 pre-existent AF; n=14 incident AF on follow-up) to age- and sex-matched controls (n=37). Electrophysiological properties of the identified autoantibody were then tested in vitro with the patch clamp technique and in vivo with an experimental mouse model of immunization. RESULTS A common autoantibody response against Kir3.4 protein was detected in patients with AF and even before the development of clinically apparent AF. Kir3.4 protein forms a heterotetramer that underlies the cardiac acetylcholine-activated inwardly rectifying K+ current, IKACh. Functional studies on human induced pluripotent stem cell-derived atrial cardiomyocytes showed that anti-Kir3.4 IgG purified from patients with AF shortened action potentials and enhanced the constitutive form of IKACh, both key mediators of AF. To establish a causal relationship, we developed a mouse model of Kir3.4 autoimmunity. Electrophysiological study in Kir3.4-immunized mice showed that Kir3.4 autoantibodies significantly reduced atrial effective refractory period and predisposed animals to a 2.8-fold increased susceptibility to AF. CONCLUSIONS To our knowledge, this is the first report of an autoimmune pathogenesis of AF with direct evidence of Kir3.4 autoantibody-mediated AF.
Collapse
Affiliation(s)
- Ange Maguy
- Institute of Physiology, University of Bern, Switzerland (A.M.)
| | | | - Jean-Claude Tardif
- Montreal Heart Institute, Université de Montréal, Canada (J.-C.T., D.B.)
| | - David Busseuil
- Montreal Heart Institute, Université de Montréal, Canada (J.-C.T., D.B.)
| | - Jin Li
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, Switzerland (J.L.)
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland (J.L.)
| |
Collapse
|
4
|
Kresovich JK, Xu Z, O'Brien KM, Parks CG, Weinberg CR, Sandler DP, Taylor JA. Peripheral Immune Cell Composition is Altered in Women Before and After a Hypertension Diagnosis. Hypertension 2023; 80:43-53. [PMID: 36259385 PMCID: PMC9742333 DOI: 10.1161/hypertensionaha.122.20001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/29/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND The development and consequences of hypertension involve multiple biological systems that may include changes in immune profiles. Whether hypertension is related to peripheral immune cell composition has not been examined in large human cohorts. METHODS We estimated circulating proportions of 12 leukocyte subsets from the lymphoid and myeloid lineages by deconvolving cell-type-specific DNA methylation data from 4124 women. Hypertension status at baseline was defined by current use of antihypertensive medication and blood pressure measurements while new incident cases were identified during follow-up via annual health questionnaires. RESULTS Among hypertension-free women at baseline, higher B cell and lower naïve CD4+ helper T cell proportions were associated with subsequent increased hazard of hypertension incidence (B cells; adjusted HR: 1.17 [95% CI: 1.02-1.35]; P=0.03; naïve CD4+ T cell, adjusted HR: 0.88 [95% CI: 0.78-0.99]; P=0.04). Blood pressure measurements at baseline were similarly positively associated with B cells and inversely associated with naïve CD4+ helper T cells. Compared to normotensive women, women with hypertension had higher circulating proportions of neutrophils (adjusted odds ratio: 1.18 [95% CI: 1.07-1.31]; P=0.001) and lower proportions of CD4+ helper T cells (adjusted odds ratio: 0.90 [95% CI: 0.81-1.00] P=0.05), natural killers (adjusted odds ratio: 0.82 [95% CI: 0.74-0.91]; P<0.001), and B cells (adjusted odds ratio: 0.84 [95% CI: 0.74-0.96]; P=0.01). CONCLUSIONS These observations suggest that shifts in lymphocyte subsets occur before hypertension development, followed by later changes to neutrophils and additional lymphocytes.
Collapse
Affiliation(s)
- Jacob K Kresovich
- Departments of Cancer Epidemiology and Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL (J.K.K.)
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC (J.K.K., Z.X., K.M.O., C.G.P., D.P.S., J.A.T.)
| | - Zongli Xu
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC (J.K.K., Z.X., K.M.O., C.G.P., D.P.S., J.A.T.)
| | - Katie M O'Brien
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC (J.K.K., Z.X., K.M.O., C.G.P., D.P.S., J.A.T.)
| | - Christine G Parks
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC (J.K.K., Z.X., K.M.O., C.G.P., D.P.S., J.A.T.)
| | - Clarice R Weinberg
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC (C.R.W.)
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC (J.K.K., Z.X., K.M.O., C.G.P., D.P.S., J.A.T.)
| | - Jack A Taylor
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC (J.K.K., Z.X., K.M.O., C.G.P., D.P.S., J.A.T.)
- Epigenetic and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC (J.A.T.)
| |
Collapse
|
5
|
Chen J, Wen Y, Chu X, Liu Y, Su C. Pulmonary adverse events associated with hypertension in non-small cell lung cancer patients receiving PD-1/PD-L1 inhibitors. Front Pharmacol 2022; 13:944342. [PMID: 36110543 PMCID: PMC9468816 DOI: 10.3389/fphar.2022.944342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 08/04/2022] [Indexed: 12/19/2022] Open
Abstract
Introduction: Non-small cell lung cancer patients have gained therapeutic benefits from immune checkpoint inhibitors, although immune-related adverse events (irAEs) could be inevitable. Whether irAEs are associated with chronic diseases is still unclear, our study aims to clarify the distinct adverse events in NSCLC patients with concomitant hypertension. Methods: Adverse event cases were searched and collected in the Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) database from January 2015 to December 2021. We performed disproportionality analysis to detect safety signals by calculating reporting odds ratios (ROR) and corresponding 95% confidence intervals (95% CIs), information component (IC), and the lower bound of the information component 95% credibility interval (IC025). Results: Among 17,163 NSCLC patients under treatment with single-agent anti-programmed death-1/programmed death ligand-1 (PD-1/PD-L1) inhibitor (nivolumab, pembrolizumab, cemiplimab, durvalumab, atezolizumab, and avelumab), 497 patients had hypertension while 16,666 patients had no hypertension. 4,283 pulmonary AEs were reported, including 166 patients with hypertension and 4,117 patients without hypertension. Compared with patients without hypertension, patients with hypertension were positively associated with increased reporting of interstitial lung disease (ROR = 3.62, 95%CI 2.68-4.89, IC = 1.54, IC025 = 0.57) among patients receiving anti-PD-1 treatment. The median duration of onset from the time of initiation of anti-PD-1 administration was 28 days (IQR, 12.00-84.25). Conclusion: Our pharmacovigilance analysis showed the profile of pulmonary toxicities in NSCLC patients with hypertension caused by anti-PD-1/PD-L1 inhibitors. Interstitial lung disease was the statistically significant reporting adverse event in patients with hypertension receiving anti-PD-1 treatment.
Collapse
Affiliation(s)
- Jianing Chen
- School of Medicine, Tongji University, Shanghai, China
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University, School of Medicine, Shanghai, China
| | - Yaokai Wen
- School of Medicine, Tongji University, Shanghai, China
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University, School of Medicine, Shanghai, China
| | - Xiangling Chu
- School of Medicine, Tongji University, Shanghai, China
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University, School of Medicine, Shanghai, China
| | - Yuzhi Liu
- School of Medicine, Tongji University, Shanghai, China
- Department of Oncology, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Chunxia Su
- School of Medicine, Tongji University, Shanghai, China
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University, School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Fan J, Wang S, Chen K, Sun Z. Aging impairs arterial compliance via Klotho-mediated downregulation of B-cell population and IgG levels. Cell Mol Life Sci 2022; 79:494. [PMID: 36001158 PMCID: PMC10082671 DOI: 10.1007/s00018-022-04512-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Aging is associated with compromised immune function and arterial remodeling and stiffness. The purpose of this study is to investigate whether in vivo AAV-based delivery of secreted Klotho (SKL) gene (AAV-SKL) improves aging- and senescence-associated immune dysfunction and arterial stiffness. METHODS AND RESULTS Senescence-accelerated mice prone strain 1 (SAMP1, 10 months) and old mice (20 months) were used. Serum SKL levels, B-cell population and serum IgG levels were markedly decreased in SAMP1 and old mice. Rescue of downregulation of serum SKL levels by in vivo AAV2-based delivery of SKL gene (AAV-SKL) increased B-cell population and serum IgG levels and attenuated arterial stiffness in SAMP1 and old mice. Thus, Klotho deficiency may play a role in senescence- and aging-associated humoral immune dysfunction and arterial stiffness. Vascular infiltration of inflammatory cells and expression of TGFβ1, collagen 1, scleraxis, MMP-2 and MMP-9 were increased while the elastin level was decreased in aortas of SAMP1 and old mice which can be rescued by AAV-SKL. Interestingly, treatment with IgG effectively rescued arterial inflammation and remodeling and attenuated arterial stiffness and hypertension in aging mice. In cultured B-lymphoblast cells, we further showed that SKL regulates B-cell proliferation and maturation partly via the NFkB pathway. CONCLUSION Aging-associated arterial stiffening may be largely attributed to downregulation of B-cell population and serum IgG levels. AAV-SKL attenuates arterial stiffness in aging mice partly via restoring B-cell population and serum IgG levels which attenuates aging-associated vascular inflammation and arterial remodeling.
Collapse
Affiliation(s)
- Jun Fan
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Shirley Wang
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Kai Chen
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, A302 Coleman Building, 956 Court Avenue, Memphis, TN, 38163, USA
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK, USA.
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, A302 Coleman Building, 956 Court Avenue, Memphis, TN, 38163, USA.
| |
Collapse
|
7
|
Jiang Y, Duffy F, Hadlock J, Raappana A, Styrchak S, Beck I, Mast FD, Miller LR, Chour W, Houck J, Armistead B, Duvvuri VR, Yeung W, Haglund M, Wallner J, Wallick JA, Hardy S, Oldroyd A, Ko D, Gervassi A, Murray KM, Kaplan H, Aitchison JD, Heath JR, Sather DN, Goldman JD, Frenkel L, Harrington WE. Angiotensin II receptor I auto-antibodies following SARS-CoV-2 infection. PLoS One 2021; 16:e0259902. [PMID: 34788328 PMCID: PMC8598062 DOI: 10.1371/journal.pone.0259902] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/28/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is associated with endothelial activation and coagulopathy, which may be related to pre-existing or infection-induced pro-thrombotic autoantibodies such as those targeting angiotensin II type I receptor (AT1R-Ab). METHODS We compared prevalence and levels of AT1R-Ab in COVID-19 cases with mild or severe disease to age and sex matched negative controls utilizing multivariate logistic and quantile regression adjusted for comorbidities including hypertension, diabetes, and heart disease. RESULTS There were trends toward increased prevalence (50% vs. 33%, p = 0.1) and level of AT1R-Ab (median 9.8 vs. 6.1 U/mL, p = 0.06) in all cases versus controls. When considered by COVID-19 disease severity, there was a trend toward increased prevalence of AT1R-Ab (55% vs. 31%, p = 0.07), as well as significantly higher AT1R-Ab levels (median 10.7 vs. 5.9 U/mL, p = 0.03) amongst individuals with mild COVID-19 versus matched controls. In contrast, the prevalence (42% vs. 37%, p = 0.9) and level (both medians 6.7 U/mL, p = 0.9) of AT1R-Ab amongst those with severe COVID-19 did not differ from matched controls. CONCLUSIONS These findings support an association between COVID-19 and AT1R-Ab, emphasizing that vascular pathology may be present in individuals with mild COVID-19 as well as those with severe disease.
Collapse
Affiliation(s)
- Yonghou Jiang
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Fergal Duffy
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Jennifer Hadlock
- Institute for Systems Biology, Seattle, Washington, United States of America
- Providence St. Joseph Health, Renton, Washington, United States of America
| | - Andrew Raappana
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Sheila Styrchak
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Ingrid Beck
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Fred D. Mast
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Leslie R. Miller
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - William Chour
- Institute for Systems Biology, Seattle, Washington, United States of America
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - John Houck
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Blair Armistead
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Venkata R. Duvvuri
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Winnie Yeung
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Micaela Haglund
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Jackson Wallner
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Julie A. Wallick
- Providence St. Joseph Health, Renton, Washington, United States of America
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, Washington, United States of America
| | - Samantha Hardy
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Alyssa Oldroyd
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Daisy Ko
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Ana Gervassi
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Kim M. Murray
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Henry Kaplan
- Swedish Cancer Institute, Swedish Medical Center, Seattle, Washington, United States of America
| | - John D. Aitchison
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - James R. Heath
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - D. Noah Sather
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Jason D. Goldman
- Providence St. Joseph Health, Renton, Washington, United States of America
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, Washington, United States of America
- Division of Allergy & Infectious Diseases, University of Washington, Seattle, Washington, United States of America
| | - Lisa Frenkel
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Whitney E. Harrington
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
8
|
Das UN. Molecular biochemical aspects of salt (sodium chloride) in inflammation and immune response with reference to hypertension and type 2 diabetes mellitus. Lipids Health Dis 2021; 20:83. [PMID: 34334139 PMCID: PMC8327432 DOI: 10.1186/s12944-021-01507-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 07/14/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity, insulin resistance, type 2 diabetes mellitus (T2DM) and hypertension (HTN) are common that are associated with low-grade systemic inflammation. Diet, genetic factors, inflammation, and immunocytes and their cytokines play a role in their pathobiology. But the exact role of sodium, potassium, magnesium and other minerals, trace elements and vitamins in the pathogenesis of HTN and T2DM is not known. Recent studies showed that sodium and potassium can modulate oxidative stress, inflammation, alter the autonomic nervous system and induce dysfunction of the innate and adaptive immune responses in addition to their action on renin-angiotensin-aldosterone system. These actions of sodium, potassium and magnesium and other minerals, trace elements and vitamins are likely to be secondary to their action on pro-inflammatory cytokines IL-6, TNF-α and IL-17 and metabolism of essential fatty acids that may account for their involvement in the pathobiology of insulin resistance, T2DM, HTN and autoimmune diseases.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 2221 NW 5th St, Battle Ground, WA, 98604, USA.
| |
Collapse
|
9
|
Chen Y, Dale BL, Alexander MR, Xiao L, Ao M, Pandey AK, Smart CD, Davis GK, Madhur MS. Class switching and high-affinity immunoglobulin G production by B cells is dispensable for the development of hypertension in mice. Cardiovasc Res 2021; 117:1217-1228. [PMID: 32609312 PMCID: PMC7983008 DOI: 10.1093/cvr/cvaa187] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/15/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS Elevated serum immunoglobulins have been associated with experimental and human hypertension for decades but whether immunoglobulins and B cells play a causal role in hypertension pathology is unclear. In this study, we sought to determine the role of B cells and high-affinity class-switched immunoglobulins on hypertension and hypertensive end-organ damage to determine if they might represent viable therapeutic targets for this disease. METHODS AND RESULTS We purified serum immunoglobulin G (IgG) from mice exposed to vehicle or angiotensin (Ang) II to induce hypertension and adoptively transferred these to wild type (WT) recipient mice receiving a subpressor dose of Ang II. We found that transfer of IgG from hypertensive animals does not affect blood pressure, endothelial function, renal inflammation, albuminuria, or T cell-derived cytokine production compared with transfer of IgG from vehicle infused animals. As an alternative approach to investigate the role of high-affinity, class-switched immunoglobulins, we studied mice with genetic deletion of activation-induced deaminase (Aicda-/-). These mice have elevated levels of IgM but virtual absence of class-switched immunoglobulins such as IgG subclasses and IgA. Neither male nor female Aicda-/- mice were protected from Ang II-induced hypertension and renal/vascular damage. To determine if IgM or non-immunoglobulin-dependent innate functions of B cells play a role in hypertension, we studied mice with severe global B-cell deficiency due to deletion of the membrane exon of the IgM heavy chain (µMT-/-). µMT-/- mice were also not protected from hypertension or end-organ damage induced by Ang II infusion or deoxycorticosterone acetate-salt treatment. CONCLUSIONS These results suggest that B cells and serum immunoglobulins do not play a causal role in hypertension pathology.
Collapse
Affiliation(s)
- Yuhan Chen
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Bethany L Dale
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Matthew R Alexander
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Liang Xiao
- Department of Medicine, Division of Clinical Pharmacology, VUMC, 2215 Garland Avenue, P415D MRB IV, Nashville, TN 37232, USA
| | - Mingfang Ao
- Department of Medicine, Division of Clinical Pharmacology, VUMC, 2215 Garland Avenue, P415D MRB IV, Nashville, TN 37232, USA
| | - Arvind K Pandey
- Department of Medicine, Division of Clinical Pharmacology, VUMC, 2215 Garland Avenue, P415D MRB IV, Nashville, TN 37232, USA
| | - Charles D Smart
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Gwendolyn K Davis
- Department of Medicine, Division of Clinical Pharmacology, VUMC, 2215 Garland Avenue, P415D MRB IV, Nashville, TN 37232, USA
| | - Meena S Madhur
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Medicine, Division of Clinical Pharmacology, VUMC, 2215 Garland Avenue, P415D MRB IV, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, USA
| |
Collapse
|
10
|
Abuzeineh M, Kyeso Y, Philogene MC, Alachkar N, Alasfar S. Presentation and Outcomes of Antibody-Mediated Rejection Associated With Angiotensin II Receptor 1 Antibodies Among Kidney Transplant Recipients. Transplant Proc 2021; 53:1501-1508. [PMID: 33573814 DOI: 10.1016/j.transproceed.2021.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/30/2020] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND It remains challenging to manage antibody-mediated rejection (ABMR) associated with angiotensin II type 1 receptor antibodies (AT1R-Abs) in kidney transplant recipients and the outcomes are not well defined. We describe the presentation, clinical course, and outcomes of this condition. METHODS This retrospective study included kidney transplant recipients with AT1R-Ab levels ≥10 units/mL and biopsy-proven ABMR in the absence of significant HLA-donor-specific antibodies at the time of rejection. RESULTS We identified 13 recipients. Median creatinine (Cr) at rejection was significantly higher (2.05 mg/dL) compared with baseline (1.2 mg/dL), P = .006. After ABMR management, the difference in median Cr was not significant (1.5 mg/dL), P = .152. Median AT1R-Ab level was higher in the pretransplant sample (34.5 units/mL) compared with the level at rejection (19 units/mL) and after rejection treatment (13 units/mL); however, these differences were not significant, P = .129. Eight of the 13 recipients received antibody reduction therapy with plasmapheresis and intravenous immunoglobulin, and 5 of the 13 recipients had other therapies. After rejection management, 6 of the 13 recipients had improvement in Cr to baseline and 7 of the 13 recipients had > 50% reduction in proteinuria. CONCLUSIONS AT1R-Ab-associated ABMR management and outcomes depend on the clinical presentation and may include antibody-reducing therapies among other therapies. Further prospective cohorts will improve recognizing and managing this condition.
Collapse
Affiliation(s)
- Mohammad Abuzeineh
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yousuf Kyeso
- Department of Medicine, University of Chicago, Chicago, Illinois
| | | | - Nada Alachkar
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sami Alasfar
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
11
|
McClung DM, Kalusche WJ, Jones KE, Ryan MJ, Taylor EB. Hypertension and endothelial dysfunction in the pristane model of systemic lupus erythematosus. Physiol Rep 2021; 9:e14734. [PMID: 33527772 PMCID: PMC7851437 DOI: 10.14814/phy2.14734] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 12/19/2020] [Indexed: 01/01/2023] Open
Abstract
Autoimmune diseases such as psoriasis, rheumatoid arthritis, and systemic lupus erythematosus (SLE) have high rates of hypertension and cardiovascular disease. Systemic lupus erythematosus is a prototypic autoimmune disorder that primarily affects women of childbearing age and is associated with a loss of self-tolerance, autoreactive B and T lymphocytes, and the production of autoantibodies, especially to nuclear components. In this study, we hypothesized that the pristane-inducible model of SLE would develop hypertension and vascular dysfunction as the disease progressed. To test this hypothesis, female C57BL/6 mice were administered PBS or pristane. Seven months after pristane administration, mice developed various autoantibodies, including anti-dsDNA IgG, anti-ssDNA IgG, and anti-nRNP IgG, as well as hypergammaglobulinemia. Several other immunological changes, including increased circulating neutrophils and increased CD4- CD8- (double negative) thymocytes were also detected. Mean arterial pressure (MAP) was elevated in pristane-treated mice when compared to PBS-treated mice. In addition, second-order mesenteric arteries from pristine-treated mice had impaired relaxation to the endothelium-dependent vasodilator acetylcholine compared to PBS-treated mice. These data suggest that the immune system dysfunction present in the pristane model of lupus contributes to the development of hypertension and vascular dysfunction.
Collapse
Affiliation(s)
- Daniel M. McClung
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - William J. Kalusche
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Katie E. Jones
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Michael J. Ryan
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
- G.V. (Sonny) Montgomery Veterans Affairs Medical CenterJacksonMSUSA
| | - Erin B. Taylor
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Inflammatory processes play a critical role in the pathogenesis of hypertension. Innate and adaptive immune responses participate in blood pressure (BP) elevation and end-organ damage. In this review, we discuss recent studies illustrating mechanisms through which immune cells and cytokines regulate BP via their actions in the kidney. RECENT FINDINGS Cells of the innate immune system, including monocytes, neutrophils, and dendritic cells, can all promote BP elevation via effects on kidney function. These innate immune cells can directly impact oxidative stress and cytokine generation in the kidney and/or present antigens to lymphocytes for the engagement of the adaptive immune system. Once activated by dendritic cells, effector memory T cells accumulate in the hypertensive kidney and facilitate renal salt and water retention. Individual subsets of activated T cells can secrete tumor necrosis factor-alpha (TNF-α), interleukin-17a (IL-17a), and interferon-gamma (IFN-γ), each of which has augmented the elevation of blood pressure in hypertensive models by enhancing renal sodium transport. B cells, regulate blood pressure via vasopressin receptor 2 (V2R)-dependent effects on fluid transport in the kidney. SUMMARY Immune cells of the innate and adaptive immune systems drive sodium retention and blood pressure elevation in part by altering renal solute transport.
Collapse
|
13
|
IgGs from patients with amyotrophic lateral sclerosis and diabetes target Ca Vα 2δ1 subunits impairing islet cell function and survival. Proc Natl Acad Sci U S A 2019; 116:26816-26822. [PMID: 31826954 PMCID: PMC6936400 DOI: 10.1073/pnas.1911956116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We provide evidence of a mechanistic link between ALS and T2DM. Our data show that a subgroup of ALS-T2DM patients have sera that enhance CaV1 channel-mediated Ca2+ influx and exaggerate [Ca2+]i. These effects occur because the sera accommodate cytotoxic IgG autoantibodies that immunocapture CaVα2δ1 subunits. As a consequence, impairments in [Ca2+]i dynamics, mitochondrial function, insulin secretion, and cell viability appear. We could clarify not only the identity of this serum factor but also the molecular mechanisms underlying its effects on the islet cells. Our findings may lay the foundation for a treatment strategy for this complex and severe group of diabetic patients. Patients with amyotrophic lateral sclerosis (ALS) often show hallmarks of type 2 diabetes mellitus (T2DM). However, the causal link between ALS and T2DM has remained a mystery. We now demonstrate that 60% of ALS patients with T2DM (ALS-T2DM) have sera that exaggerated K+-induced increases in cytosolic free Ca2+ concentration ([Ca2+]i) in mouse islet cells. The effect was attributed to the presence of pathogenic immunoglobulin Gs (IgGs) in ALS-T2DM sera. The pathogenic IgGs immunocaptured the voltage-dependent Ca2+ (CaV) channel subunit CaVα2δ1 in the plasma membrane enhancing CaV1 channel-mediated Ca2+ influx and [Ca2+]i, resulting in impaired mitochondrial function. Consequently, impairments in [Ca2+]i dynamics, insulin secretion, and cell viability occurred. These data reveal that patients with ALS-T2DM carry cytotoxic ALS-T2DM-IgG autoantibodies that serve as a causal link between ALS and T2DM by immunoattacking CaVα2δ1 subunits. Our findings may lay the foundation for a pharmacological treatment strategy for patients suffering from a combination of these diseases.
Collapse
|
14
|
Peng J, Vongpatanasin W, Sacharidou A, Kifer D, Yuhanna IS, Banerjee S, Tanigaki K, Polasek O, Chu H, Sundgren NC, Rohatgi A, Chambliss KL, Lauc G, Mineo C, Shaul PW. Supplementation With the Sialic Acid Precursor N-Acetyl-D-Mannosamine Breaks the Link Between Obesity and Hypertension. Circulation 2019; 140:2005-2018. [PMID: 31597453 PMCID: PMC7027951 DOI: 10.1161/circulationaha.119.043490] [Citation(s) in RCA: 39] [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] [Indexed: 12/21/2022]
Abstract
BACKGROUND Obesity-related hypertension is a common disorder, and attempts to combat the underlying obesity are often unsuccessful. We previously revealed that mice globally deficient in the inhibitory immunoglobulin G (IgG) receptor FcγRIIB are protected from obesity-induced hypertension. However, how FcγRIIB participates is unknown. Studies were designed to determine if alterations in IgG contribute to the pathogenesis of obesity-induced hypertension. METHODS Involvement of IgG was studied using IgG μ heavy chain-null mice deficient in mature B cells and by IgG transfer. Participation of FcγRIIB was interrogated in mice with global or endothelial cell-specific deletion of the receptor. Obesity was induced by high-fat diet (HFD), and blood pressure (BP) was measured by radiotelemetry or tail cuff. The relative sialylation of the Fc glycan on mouse IgG, which influences IgG activation of Fc receptors, was evaluated by Sambucus nigra lectin blotting. Effects of IgG on endothelial NO synthase were assessed in human aortic endothelial cells. IgG Fc glycan sialylation was interrogated in 3442 human participants by mass spectrometry, and the relationship between sialylation and BP was evaluated. Effects of normalizing IgG sialylation were determined in HFD-fed mice administered the sialic acid precursor N-acetyl-D-mannosamine (ManNAc). RESULTS Mice deficient in B cells were protected from obesity-induced hypertension. Compared with IgG from control chow-fed mice, IgG from HFD-fed mice was hyposialylated, and it raised BP when transferred to recipients lacking IgG; the hypertensive response was absent if recipients were FcγRIIB-deficient. Neuraminidase-treated IgG lacking the Fc glycan terminal sialic acid also raised BP. In cultured endothelial cells, via FcγRIIB, IgG from HFD-fed mice and neuraminidase-treated IgG inhibited vascular endothelial growth factor activation of endothelial NO synthase by altering endothelial NO synthase phosphorylation. In humans, obesity was associated with lower IgG sialylation, and systolic BP was inversely related to IgG sialylation. Mice deficient in FcγRIIB in endothelium were protected from obesity-induced hypertension. Furthermore, in HFD-fed mice, ManNAc normalized IgG sialylation and prevented obesity-induced hypertension. CONCLUSIONS Hyposialylated IgG and FcγRIIB in endothelium are critically involved in obesity-induced hypertension in mice, and supportive evidence was obtained in humans. Interventions targeting these mechanisms, such as ManNAc supplementation, may provide novel means to break the link between obesity and hypertension.
Collapse
Affiliation(s)
- Jun Peng
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| | - Wanpen Vongpatanasin
- Division of Cardiology, Dept. of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| | - Anastasia Sacharidou
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| | - Domagoj Kifer
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Ivan S. Yuhanna
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| | - Subhashis Banerjee
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| | - Keiji Tanigaki
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| | - Ozren Polasek
- Department of Public Health, University of Split School of Medicine, Split, Croatia
| | - Haiyan Chu
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| | - Nathan C. Sundgren
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| | - Anand Rohatgi
- Division of Cardiology, Dept. of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| | - Ken L. Chambliss
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| | - Gordan Lauc
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| | - Philip W. Shaul
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX USA 75390
| |
Collapse
|
15
|
Dale BL, Pandey AK, Chen Y, Smart CD, Laroumanie F, Ao M, Xiao L, Dikalova AE, Dikalov SI, Elijovich F, Foss JD, Barbaro NR, Van Beusecum JP, Deger SM, Alsouqi A, Itani HA, Norlander AE, Alexander MR, Zhao S, Ikizler TA, Algood HMS, Madhur MS. Critical role of Interleukin 21 and T follicular helper cells in hypertension and vascular dysfunction. JCI Insight 2019; 5:129278. [PMID: 31013256 DOI: 10.1172/jci.insight.129278] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
T and B cells have been implicated in hypertension, but the mechanisms by which they produce a coordinated response is unknown. T follicular helper (Tfh) cells that produce interleukin 21 (IL21) promote germinal center (GC) B cell responses leading to immunoglobulin (Ig) production. Here we investigate the role of IL21 and Tfh cells in hypertension. In response to angiotensin (Ang) II-induced hypertension, T cell IL21 production is increased, and Il21-/- mice develop blunted hypertension, attenuated vascular end-organ damage, and decreased interleukin 17A (IL17A) and interferon gamma production. Tfh-like cells and GC B cells accumulate in the aorta and plasma IgG1 is increased in hypertensive WT but not Il21-/-mice. Furthermore, Tfh cell deficient mice develop blunted hypertension and vascular hypertrophy in response to Ang II infusion. Importantly, IL21 neutralization reduces blood pressure (BP) and reverses endothelial dysfunction and vascular inflammation. Moreover, recombinant IL21 impairs endothelium-dependent relaxation ex vivo and decreases nitric oxide production from cultured endothelial cells. Finally, we show in humans that peripheral blood T cell production of IL21 correlates with systolic BP and IL17A production. These data suggest that IL21 may be a novel therapeutic target for the treatment of hypertension and its micro- and macrovascular complications.
Collapse
Affiliation(s)
- Bethany L Dale
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Yuhan Chen
- Department of Medicine, Division of Clinical Pharmacology
| | - Charles D Smart
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Mingfang Ao
- Department of Medicine, Division of Clinical Pharmacology
| | - Liang Xiao
- Department of Medicine, Division of Clinical Pharmacology
| | | | | | | | - Jason D Foss
- Department of Medicine, Division of Clinical Pharmacology
| | | | | | | | | | - Hana A Itani
- Department of Medicine, Division of Clinical Pharmacology
| | - Allison E Norlander
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | | | | | | | - Holly M Scott Algood
- Department of Medicine, Division of Infectious Disease, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA.,Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee, USA.,Vanderbilt Digestive Diseases Research Center, Nashville, Tennessee, USA
| | - Meena S Madhur
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA.,Department of Medicine, Division of Cardiovascular Medicine.,Department of Medicine, Division of Clinical Pharmacology.,Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee, USA
| |
Collapse
|
16
|
Don-Doncow N, Zhang Y, Matuskova H, Meissner A. The emerging alliance of sphingosine-1-phosphate signalling and immune cells: from basic mechanisms to implications in hypertension. Br J Pharmacol 2018; 176:1989-2001. [PMID: 29856066 DOI: 10.1111/bph.14381] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/24/2018] [Accepted: 05/28/2018] [Indexed: 01/19/2023] Open
Abstract
The immune system plays a considerable role in hypertension. In particular, T-lymphocytes are recognized as important players in its pathogenesis. Despite substantial experimental efforts, the molecular mechanisms underlying the nature of T-cell activation contributing to an onset of hypertension or disease perpetuation are still elusive. Amongst other cell types, lymphocytes express distinct profiles of GPCRs for sphingosine-1-phosphate (S1P) - a bioactive phospholipid that is involved in many critical cell processes and most importantly majorly regulates T-cell development, lymphocyte recirculation, tissue-homing patterns and chemotactic responses. Recent findings have revealed a key role for S1P chemotaxis and T-cell mobilization for the onset of experimental hypertension, and elevated circulating S1P levels have been linked to several inflammation-associated diseases including hypertension in patients. In this article, we review the recent progress towards understanding how S1P and its receptors regulate immune cell trafficking and function and its potential relevance for the pathophysiology of hypertension. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
| | - Yun Zhang
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Hana Matuskova
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden.,Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Anja Meissner
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
17
|
Autoantibodies against AT1 and α1-adrenergic receptors predict arterial stiffness progression in normotensive subjects over a 5-year period. Clin Sci (Lond) 2017; 131:2947-2957. [PMID: 29097625 DOI: 10.1042/cs20171305] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/24/2017] [Accepted: 11/01/2017] [Indexed: 01/08/2023]
Abstract
Arterial stiffness is an independent indicator of cardiovascular risk. Autoantibodies (AAs) against angiotensin AT1 receptor (AT1-AAs) and α1-adrenergic receptor (α1-AAs) are important in the pathogenesis of hypertension. We identified the types of AT1-AAs and α1-AAs in normotensive subjects, with the aim of determining whether these antibodies predict aortic stiffness progression. Carotid–femoral pulse wave velocity (cf-PWV) was used to measure aortic stiffness. Overall, 816 subjects (71% of those invited) underwent a medical examination and evaluation of aortic stiffness. The types of AT1-AAs and α1-AAs were measured at baseline. Meanwhile, plasma renin, angiotensin II (Ang II), and norepinephrine (NE) concentrations were measured at baseline and follow-up. Baseline mean cf-PWV was 9.90 ± 0.84 m/s and follow-up was 10.51 ± 1.12 m/s. The annualized ΔPWV was 0.12 ± 0.08 m/s/year. At the end of follow-up, 129 normotensive subjects developed hypertension and 144 subjects had PWV progression. After adjustment for covariates, AA type was independently associated with ΔPWV, annualized ΔPWV, and abnormal PWV. In our study, the risk of developing hypertension (RR =2.028, 95% CI: 1.227–3.351, P=0.006) and PWV progression (RR =2.910, 95% CI: 1.612–5.253, P<0.001) in AA-positive subjects was significantly higher than that in AA-negative subjects. Receiver operating characteristic (ROC) curve showed AA had an identify power to discriminate subjects with or without PWV and hypertension progression. We have shown for the first time that the types of A1-AAs and α1-AAs are independent predictors for aortic stiffness progression in normotensive subjects. Our data collectively support the utility of these AAs as potential markers of aortic stiffness.
Collapse
|
18
|
Chan CT, Lieu M, Sobey CG, Drummond GR, Vinh A. Diagnosing and Treating Hypertensive Disorders of Pregnancy? Hypertension 2017; 70:884-886. [DOI: 10.1161/hypertensionaha.117.09849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Christopher T. Chan
- From the Massachusetts General Hospital, Harvard Medical School, Boston (C.T.C.); Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia (M.L.); and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia (C.G.S., G.R.D., A.V.)
| | - Maggie Lieu
- From the Massachusetts General Hospital, Harvard Medical School, Boston (C.T.C.); Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia (M.L.); and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia (C.G.S., G.R.D., A.V.)
| | - Christopher G. Sobey
- From the Massachusetts General Hospital, Harvard Medical School, Boston (C.T.C.); Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia (M.L.); and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia (C.G.S., G.R.D., A.V.)
| | - Grant R. Drummond
- From the Massachusetts General Hospital, Harvard Medical School, Boston (C.T.C.); Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia (M.L.); and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia (C.G.S., G.R.D., A.V.)
| | - Antony Vinh
- From the Massachusetts General Hospital, Harvard Medical School, Boston (C.T.C.); Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia (M.L.); and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia (C.G.S., G.R.D., A.V.)
| |
Collapse
|
19
|
Nosalski R, McGinnigle E, Siedlinski M, Guzik TJ. Novel Immune Mechanisms in Hypertension and Cardiovascular Risk. CURRENT CARDIOVASCULAR RISK REPORTS 2017; 11:12. [PMID: 28360962 PMCID: PMC5339316 DOI: 10.1007/s12170-017-0537-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Hypertension is a common disorder with substantial impact on public health due to highly elevated cardiovascular risk. The mechanisms still remain unclear and treatments are not sufficient to reduce risk in majority of patients. Inflammatory mechanisms may provide an important mechanism linking hypertension and cardiovascular risk. We aim to review newly identified immune and inflammatory mechanisms of hypertension with focus on their potential therapeutic impact. RECENT FINDINGS In addition to the established role of the vasculature, kidneys and central nervous system in pathogenesis of hypertension, low-grade inflammation contributes to this disorder as indicated by experimental models and GWAS studies pointing to SH2B3 immune gene as top key driver of hypertension. Immune responses in hypertension are greatly driven by neoantigens generated by oxidative stress and modulated by chemokines such as RANTES, IP-10 and microRNAs including miR-21 and miR-155 with other molecules under investigation. Cells of both innate and adoptive immune system infiltrate vasculature and kidneys, affecting their function by releasing pro-inflammatory mediators and reactive oxygen species. SUMMARY Immune and inflammatory mechanisms of hypertension provide a link between high blood pressure and increased cardiovascular risk, and reduction of blood pressure without attention to these underlying mechanisms is not sufficient to reduce risk.
Collapse
Affiliation(s)
- Ryszard Nosalski
- BHF Centre for Excellence Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland UK
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Eilidh McGinnigle
- BHF Centre for Excellence Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland UK
| | - Mateusz Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz J. Guzik
- BHF Centre for Excellence Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland UK
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
20
|
Selvaraj UM, Poinsatte K, Torres V, Ortega SB, Stowe AM. Heterogeneity of B Cell Functions in Stroke-Related Risk, Prevention, Injury, and Repair. Neurotherapeutics 2016; 13:729-747. [PMID: 27492770 PMCID: PMC5081124 DOI: 10.1007/s13311-016-0460-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It is well established that post-stroke inflammation contributes to neurovascular injury, blood-brain barrier disruption, and poor functional recovery in both animal and clinical studies. However, recent studies also suggest that several leukocyte subsets, activated during the post-stroke immune response, can exhibit both pro-injury and pro-recovery phenotypes. In accordance with these findings, B lymphocytes, or B cells, play a heterogeneous role in the adaptive immune response to stroke. This review highlights what is currently understood about the various roles of B cells, with an emphasis on stroke risk factors, as well as post-stroke injury and repair. This includes an overview of B cell functions, such as antibody production, cytokine secretion, and contribution to the immune response as antigen presenting cells. Next, evidence for B cell-mediated mechanisms in stroke-related risk factors, including hypertension, diabetes, and atherosclerosis, is outlined, followed by studies that focus on B cells during endogenous protection from stroke. Subsequently, animal studies that investigate the role of B cells in post-stroke injury and repair are summarized, and the final section describes current B cell-related clinical trials for stroke, as well as other central nervous system diseases. This review reveals the complex role of B cells in stroke, with a focus on areas for potential clinical intervention for a disease that affects millions of people globally each year.
Collapse
Affiliation(s)
- Uma Maheswari Selvaraj
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Katherine Poinsatte
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Vanessa Torres
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Sterling B Ortega
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Ann M Stowe
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA.
| |
Collapse
|
21
|
Khamis RY, Hughes AD, Caga-Anan M, Chang CL, Boyle JJ, Kojima C, Welsh P, Sattar N, Johns M, Sever P, Mayet J, Haskard DO. High Serum Immunoglobulin G and M Levels Predict Freedom From Adverse Cardiovascular Events in Hypertension: A Nested Case-Control Substudy of the Anglo-Scandinavian Cardiac Outcomes Trial. EBioMedicine 2016; 9:372-380. [PMID: 27333022 PMCID: PMC4972545 DOI: 10.1016/j.ebiom.2016.06.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/20/2016] [Accepted: 06/06/2016] [Indexed: 10/25/2022] Open
Abstract
AIMS We aimed to determine whether the levels of total serum IgM and IgG, together with specific antibodies against malondialdehyde-conjugated low-density lipoprotein (MDA-LDL), can improve cardiovascular risk discrimination. METHODS AND RESULTS The Anglo-Scandinavian Cardiac Outcomes Trial (ASCOT) randomized 9098 patients in the UK and Ireland into the Blood Pressure-Lowering Arm. 485 patients that had cardiovascular (CV) events over 5.5years were age and sex matched with 1367 controls. Higher baseline total serum IgG, and to a lesser extent IgM, were associated with decreased risk of CV events (IgG odds ratio (OR) per one standard deviation (SD) 0.80 [95% confidence interval, CI 0.72,0.89], p<0.0001; IgM 0.83[0.75,0.93], p=0.001), and particularly events due to coronary heart disease (CHD) (IgG OR 0.66 (0.57,0.76); p<0.0001, IgM OR 0.81 (0.71,0.93); p=0.002). The association persisted after adjustment for a basic model with variables in the Framingham Risk Score (FRS) as well as following inclusion of C-reactive protein (CRP) and N-terminal pro-B-type natriuretic peptide (NtProBNP). IgG and IgM antibodies against MDA-LDL were also associated with CV events but their significance was lost following adjustment for total serum IgG and IgM respectively. The area under the receiver operator curve for CV events was improved from the basic risk model when adding in total serum IgG, and there was improvement in continuous and categorical net reclassification (17.6% and 7.5% respectively) as well as in the integrated discrimination index. CONCLUSION High total serum IgG levels are an independent predictor of freedom from adverse cardiovascular events, particularly those attributed to CHD, in patients with hypertension.
Collapse
Affiliation(s)
- Ramzi Y Khamis
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom
| | - Alun D Hughes
- International Centre for Circulatory Health, NHLI, Imperial College London, United Kingdom; Institute of Cardiovascular Science, University College London, United Kingdom
| | - Mikhail Caga-Anan
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom
| | - Choon L Chang
- International Centre for Circulatory Health, NHLI, Imperial College London, United Kingdom
| | - Joseph J Boyle
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom
| | - Chiari Kojima
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom
| | - Paul Welsh
- Division of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Naveed Sattar
- Division of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Michael Johns
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom
| | - Peter Sever
- International Centre for Circulatory Health, NHLI, Imperial College London, United Kingdom
| | - Jamil Mayet
- International Centre for Circulatory Health, NHLI, Imperial College London, United Kingdom
| | - Dorian O Haskard
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom.
| |
Collapse
|
22
|
Chan CT, Sobey CG, Lieu M, Ferens D, Kett MM, Diep H, Kim HA, Krishnan SM, Lewis CV, Salimova E, Tipping P, Vinh A, Samuel CS, Peter K, Guzik TJ, Kyaw TS, Toh BH, Bobik A, Drummond GR. Obligatory Role for B Cells in the Development of Angiotensin II–Dependent Hypertension. Hypertension 2015; 66:1023-33. [DOI: 10.1161/hypertensionaha.115.05779] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/07/2015] [Indexed: 01/08/2023]
Abstract
Clinical hypertension is associated with raised serum IgG antibodies. However, whether antibodies are causative agents in hypertension remains unknown. We investigated whether hypertension in mice is associated with B-cell activation and IgG production and moreover whether B-cell/IgG deficiency affords protection against hypertension and vascular remodeling. Angiotensin II (Ang II) infusion (0.7 mg/kg per day; 28 days) was associated with (1) a 25% increase in the proportion of splenic B cells expressing the activation marker CD86, (2) an 80% increase in splenic plasma cell numbers, (3) a 500% increase in circulating IgG, and (4) marked IgG accumulation in the aortic adventitia. In B-cell–activating factor receptor–deficient (BAFF-R
−/−
) mice, which lack mature B cells, there was no evidence of Ang II–induced increases in serum IgG. Furthermore, the hypertensive response to Ang II was attenuated in BAFF-R
−/−
(Δ30±4 mm Hg) relative to wild-type (Δ41±5 mm Hg) mice, and this response was rescued by B-cell transfer. BAFF-R
−/−
mice displayed reduced IgG accumulation in the aorta, which was associated with 80% fewer aortic macrophages and a 70% reduction in transforming growth factor-β expression. BAFF-R
−/−
mice were also protected from Ang II–induced collagen deposition and aortic stiffening (assessed by pulse wave velocity analysis). Finally, like BAFF-R deficiency, pharmacological depletion of B cells with an anti-CD20 antibody attenuated Ang II–induced hypertension by ≈35%. Hence, these studies demonstrate that B cells/IgGs are crucial for the development of Ang II–induced hypertension and vessel remodeling in mice. Thus, B-cell–targeted therapies—currently used for autoimmune diseases—may hold promise as future treatments for hypertension.
Collapse
Affiliation(s)
- Christopher T. Chan
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Christopher G. Sobey
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Maggie Lieu
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Dorota Ferens
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Michelle M. Kett
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Henry Diep
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Hyun Ah Kim
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Shalini M. Krishnan
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Caitlin V. Lewis
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Ekaterina Salimova
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Peter Tipping
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Antony Vinh
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Chrishan S. Samuel
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Karlheinz Peter
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Tomasz J. Guzik
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Tin S. Kyaw
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Ban-Hock Toh
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Alexander Bobik
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| | - Grant R. Drummond
- From the Cardiovascular Disease Program, Biomedicine Discovery Institute (C.T.C., C.G.S., M.L., D.F., M.M.K., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Pharmacology (C.T.C., C.G.S., M.L., D.F., H.D., H.A.K., S.M.K., C.V.L., A.V., C.S.S., G.R.D.), Department of Surgery, Monash Health (C.G.S., G.R.D.), Department of Physiology (M.M.K.), Australian Regenerative Medicine Institute (E.S.), and Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School (P.T
| |
Collapse
|
23
|
Tanigaki K, Sundgren N, Khera A, Vongpatanasin W, Mineo C, Shaul PW. Fcγ receptors and ligands and cardiovascular disease. Circ Res 2015; 116:368-84. [PMID: 25593280 DOI: 10.1161/circresaha.116.302795] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Fcγ receptors (FcγRs) classically modulate intracellular signaling on binding of the Fc region of IgG in immune response cells. How FcγR and their ligands affect cardiovascular health and disease has been interrogated recently in both preclinical and clinical studies. The stimulation of activating FcγR in endothelial cells, vascular smooth muscle cells, and monocytes/macrophages causes a variety of cellular responses that may contribute to vascular disease pathogenesis. Stimulation of the lone inhibitory FγcR, FcγRIIB, also has adverse consequences in endothelial cells, antagonizing NO production and reparative mechanisms. In preclinical disease models, activating FcγRs promote atherosclerosis, whereas FcγRIIB is protective, and activating FcγRs also enhance thrombotic and nonthrombotic vascular occlusion. The FcγR ligand C-reactive protein (CRP) has undergone intense study. Although in rodents CRP does not affect atherosclerosis, it causes hypertension and insulin resistance and worsens myocardial infarction. Massive data have accumulated indicating an association between increases in circulating CRP and coronary heart disease in humans. However, Mendelian randomization studies reveal that CRP is not likely a disease mediator. CRP genetics and hypertension warrant further investigation. To date, studies of genetic variants of activating FcγRs are insufficient to implicate the receptors in coronary heart disease pathogenesis in humans. However, a link between FcγRIIB and human hypertension may be emerging. Further knowledge of the vascular biology of FcγR and their ligands will potentially enhance our understanding of cardiovascular disorders, particularly in patients whose greater predisposition for disease is not explained by traditional risk factors, such as individuals with autoimmune disorders.
Collapse
Affiliation(s)
- Keiji Tanigaki
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas
| | - Nathan Sundgren
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas
| | - Amit Khera
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas
| | - Wanpen Vongpatanasin
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas
| | - Chieko Mineo
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas
| | - Philip W Shaul
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas.
| |
Collapse
|