1
|
Xia D, Zheng Q, Liu Y, Wang L, Wei D. Targeting Immune Cell Metabolism: A Promising Therapeutic Approach for Cardiovascular Disease. Immunology 2025; 175:134-150. [PMID: 40129229 DOI: 10.1111/imm.13913] [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: 10/15/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 03/26/2025] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality globally. Recent groundbreaking preclinical and clinical research underscores the pivotal role of metabolite remodelling in the pathology of CVD. This metabolic transformation not only directly fuels the progression of CVD but also profoundly influences the immune response within the cardiovascular system. In this review, we focused on the complex interactions between cardiovascular metabolic alterations and immune responses during the course of CVD. Furthermore, we explore the potential therapeutic interventions that could be developed based on the understanding of metabolic alterations and immune dysregulation in CVD. By targeting these metabolic and immunological pathways, novel strategies for the prevention and treatment of CVDs might be developed to improve patient outcomes and reduce the global burden of this disease.
Collapse
Affiliation(s)
- Dexiang Xia
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Qinwen Zheng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Yue Liu
- Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Wang
- Hengyang Maternal and Child Health Hospital, Hengyang, China
| | - Dangheng Wei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
2
|
Rendon CJ, Watts SW, Contreras GA. PVAT adipocyte: energizing, modulating, and structuring vascular function during normotensive and hypertensive states. Am J Physiol Heart Circ Physiol 2025; 328:H1204-H1217. [PMID: 40250838 DOI: 10.1152/ajpheart.00093.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/20/2025] [Accepted: 04/11/2025] [Indexed: 04/20/2025]
Abstract
Hypertension represents the most common form of cardiovascular disease. It is characterized by significant remodeling of the various layers of the vascular system, including the outermost layer: the perivascular adipose tissue (PVAT). Given the tissue's pivotal role in regulating blood pressure, a comprehensive understanding of the changes that occur within PVAT during the progression of hypertension is essential. This article reviews the mechanisms through which PVAT modulates blood pressure, including the secretion of bioactive soluble factors, provision of mechanical support, and adipose-specific functions such as adipogenesis, lipogenesis, lipolysis, and extracellular matrix remodeling. Additionally, this review emphasizes the influence of hypertension on each of these regulatory mechanisms, thereby providing a deeper insight into the pathophysiological interplay between hypertension and PVAT biology.
Collapse
Affiliation(s)
- C Javier Rendon
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, United States
| |
Collapse
|
3
|
Berillo O, Paradis P, Schiffrin EL. Role of Immune Cells in Perivascular Adipose Tissue in Vascular Injury in Hypertension. Arterioscler Thromb Vasc Biol 2025; 45:563-575. [PMID: 40079139 DOI: 10.1161/atvbaha.124.321689] [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] [Indexed: 03/14/2025]
Abstract
Hypertension is associated with vascular injury characterized by vascular dysfunction, remodeling, and stiffening, which contributes to end-organ damage leading to cardiovascular events and potentially death. Innate (macrophages and dendritic cells), innate-like (γδ T cells) and adaptive immune cells (T and B cells) play a role in hypertension and vascular injury. Perivascular adipose tissue that is the fourth layer of the blood vessel wall is an important homeostatic regulator of vascular tone. Increased infiltration of immune cells in perivascular adipose tissue in hypertension results in generation of oxidative stress and production of cytokines that may cause vascular injury. This review presents an overview of the role of the different immune cells that infiltrate the perivascular adipose tissue and are involved in the pathophysiology of hypertension.
Collapse
Affiliation(s)
- Olga Berillo
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research, Quebec, Canada (O.B., P.P., E.L.S.)
| | - Pierre Paradis
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research, Quebec, Canada (O.B., P.P., E.L.S.)
| | - Ernesto L Schiffrin
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research, Quebec, Canada (O.B., P.P., E.L.S.)
- Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, Montréal, Québec, Canada (E.L.S.)
- McGill University, Montréal, Québec, Canada (E.L.S.)
| |
Collapse
|
4
|
Suzuki Y. Ca 2+ microdomains in vascular smooth muscle cells: Roles in vascular tone regulation and hypertension. J Pharmacol Sci 2025; 158:59-67. [PMID: 40121058 DOI: 10.1016/j.jphs.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Vascular smooth muscle cells (VSMCs) modulate blood pressure by adjusting vascular contractility. Specific families of ion channels that are expressed in VSMCs regulate membrane potential and intracellular Ca2+ concentration ([Ca2+]cyt). Subsets of them are known to form molecular complexes with Ca2+-sensitive molecules via scaffolding proteins such as caveolin and junctophilin. This enables localized and molecular complex-specific signal transduction to regulate vascular contractility. This intracellular region is referred to as a Ca2+ microdomain. When hypertensive stimuli are applied to blood vessels, gene expression of ion channels and scaffold proteins in vascular cells changes dramatically, often leading to membrane depolarization and increased [Ca2+]cyt. As a result, blood vessels undergo functional remodeling characterized by enhanced contractility. In addition, the transcription of inflammatory genes in vascular cells is also upregulated. This induces leukocyte infiltration into the vascular wall and structural remodeling mediated by VSMC proliferation and extracellular matrix remodeling. This functional and structural remodeling perpetuates the hypertensive state, leading to progressive damage to systemic organs. This review summarizes recent findings on the mechanisms by which Ca2+ microdomains in VSMCs regulate contractility. In addition, the changes in Ca2+ microdomains due to hypertensive stimuli and their contributions to both functional and structural remodeling are summarized.
Collapse
Affiliation(s)
- Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.
| |
Collapse
|
5
|
Wang X, Chai Y, Dou Y, Li X, Li F, Gao K. Global trends and hotspots in macrophage research related to hypertension from 2015-2024: bibliometric research and visualization analysis. Front Immunol 2025; 16:1501432. [PMID: 40109339 PMCID: PMC11920128 DOI: 10.3389/fimmu.2025.1501432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
Background Hypertension continues to be a global health and economic burden, conventionally characterized by a chronic inflammatory state. Macrophages are critical for the initiation, progression and manifestation of hypertension. As studies on the relationship between macrophages and hypertension increase substantially, identifying critical research areas and unraveling potential interaction mechanisms become increasingly essential. Methods Articles associated with hypertension and macrophages in recent 10 years were retrieved from the Web of Science Core Collection for analysis, using Microsoft Excel, VOSviewer, CiteSpace and Scimago Graphica. Results After excluding studies that did not meet inclusive standard based on time (2015-2024) and type (article or reviews), 2,013 original articles related to macrophages associated with hypertension were included. The number of publications has been increasing annually. These records consisted of 2,013 English language papers published in 351 journals by 315 institutions or regions from 83 countries/regions between 2015 and 2024. We analyzed the co-cited references clusters to objectively outline the current state of research, including the regulatory mechanisms of hypertension, diseases related to hypertension, and the lifestyle factor. Inflammation remains one of the most popular research hot-spot. The most popular publishing journal in this field is PLOS ONE and the most prolific writer is Li, Hui-Hua. The primary keywords cluster in this field is inflammation, with the highest occurrences and TLS among the top 10 keywords. Conclusion These comprehensive and visualized bibliometric results summarized the significant findings in macrophage-related hypertension studies over the past 10 years. Macrophages appear to be effective in the treatment of hypertension as potential targets, but further research is needed to clarify the specific pathophysiological mechanisms involved.
Collapse
Affiliation(s)
| | | | | | | | - Fanghe Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Kuo Gao
- Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Strohm L, Ubbens H, Mihalikova D, Czarnowski A, Stamm P, Molitor M, Finger S, Oelze M, Atzler D, Wenzel P, Lurz P, Münzel T, Weber C, Lutgens E, Daiber A, Daub S. CD40-TRAF6 inhibition suppresses cardiovascular inflammation, oxidative stress and functional complications in a mouse model of arterial hypertension. Redox Biol 2025; 80:103520. [PMID: 39899926 PMCID: PMC11840497 DOI: 10.1016/j.redox.2025.103520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025] Open
Abstract
Cardiovascular disease is the leading cause of disease burden and death worldwide and is fueled by vascular inflammation. CD40L-CD40-TRAF signaling is involved in the progression of atherosclerosis and drives the development of coronary heart disease (CHD). The present study investigates whether the CD40L-CD40-TRAF6 signaling pathway with focus on immune cells and adipocytes could be a therapeutic target in arterial hypertension. Arterial hypertension was induced in WT (C57BL6/J) and cell-specific CD40(L) knockout mice (AdipoqCre x CD40 fl/fl, CD4Cre x CD40 fl/fl, CD19Cre x CD40 fl/fl, and GP1baCre x CD40L fl/fl) via angiotensin (AT-II) infusion (1 mg/kg/d) for seven days. Hypertensive WT mice were also treated with a CD40-TRAF6 inhibitor (2.5 mg/kg/d, for 7d). The TRAF6 inhibitor treatment normalized endothelial dysfunction and reduced blood pressure in hypertensive wild type animals. Reactive oxygen species production was decreased by TRAF6 inhibition in blood, aorta, heart, kidney, and perivascular fat tissue. Additionally, FACS analysis revealed that TRAF6 inhibition prevents immune cell migration into the aortic vessel wall observed by reduced CD45+ leukocyte, Ly6G+/Ly6C+ neutrophil, and Ly6Chigh inflammatory monocyte content. The hypertensive cell type-specific CD40(L) knockout animals showed only a minor effect on endothelial function, blood pressure, and oxidative stress. Therefore, we conclude that targeting CD40 directly on adipocytes, B-cells, T-cells, or CD40L on platelets is not a promising target to prevent hypertension complications. In summary, TRAF6 inhibition but not adipocyte, B-cell, or T-cell-specific CD40 or platelet-specific CD40L deficiency reduces pathophysiological vascular inflammation in hypertensive mice, suggesting TRAF6 inhibition as a potential therapeutic target in hypertensive patients.
Collapse
Affiliation(s)
- Lea Strohm
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Henning Ubbens
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Dominika Mihalikova
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Alexander Czarnowski
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Paul Stamm
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Michael Molitor
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany
| | - Stefanie Finger
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Matthias Oelze
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Germany; Walter Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Philip Wenzel
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany
| | - Philipp Lurz
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Esther Lutgens
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Germany; Mayo Clinic, Dept Cardiovascular Medicine and Immunology, Rochester, MN, USA
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany.
| | - Steffen Daub
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
7
|
Dasinger JH, Abais-Battad JM, McCrorey MK, Van Beusecum JP. Recent advances on immunity and hypertension: the new cells on the kidney block. Am J Physiol Renal Physiol 2025; 328:F301-F315. [PMID: 39853324 DOI: 10.1152/ajprenal.00309.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/26/2024] [Accepted: 01/20/2025] [Indexed: 01/26/2025] Open
Abstract
Over the past 50 years, the contribution of the immune system has been identified in the development of hypertension and renal injury. Both human and experimental animal models of hypertension have demonstrated that innate and adaptive immune cells, along with their cytokines and chemokines, modulate blood pressure fluctuations and end organ renal damage. Numerous cell types of the innate immune system, specifically monocytes, macrophages, and dendritic cells, present antigenic peptides to T cells, promoting inflammation and the elevation of blood pressure. These T cells and other adaptive immune cells migrate to vascular and tubular cells of the kidney and promote end-organ fibrosis, damage, and ultimately hypertensive injury. Through the development of high-throughput screening, novel renal and immune cell subsets have been identified as possible contributors and regulators of renal injury and hypertension. In this review, we will consider classical immunological cells and their contribution to renal inflammation, and novel cell subsets, including renal stromal cells, that could potentially shed new light on renal injury and hypertension. Finally, we will discuss how interorgan inflammation contributes to the development of hypertension and hypertension-related multiorgan damage, and explore the clinical implications of the immunological components of renal injury and hypertension.
Collapse
Affiliation(s)
- John Henry Dasinger
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, United States
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Georgia, August University, Augusta, Georgia, United States
| | - Marice K McCrorey
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Justin P Van Beusecum
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
- Department of Research and Development, Ralph H. Johnson VA Healthcare System, Charleston, South Carolina, United States
| |
Collapse
|
8
|
Xia T, Yu J, Du M, Chen X, Wang C, Li R. Vascular endothelial cell injury: causes, molecular mechanisms, and treatments. MedComm (Beijing) 2025; 6:e70057. [PMID: 39931738 PMCID: PMC11809559 DOI: 10.1002/mco2.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 02/13/2025] Open
Abstract
Vascular endothelial cells form a single layer of flat cells that line the inner surface of blood vessels, extending from large vessels to the microvasculature of various organs. These cells are crucial metabolic and endocrine components of the body, playing vital roles in maintaining circulatory stability, regulating vascular tone, and preventing coagulation and thrombosis. Endothelial cell injury is regarded as a pivotal initiating factor in the pathogenesis of various diseases, triggered by multiple factors, including infection, inflammation, and hemodynamic changes, which significantly compromise vascular integrity and function. This review examines the causes, underlying molecular mechanisms, and potential therapeutic approaches for endothelial cell injury, focusing specifically on endothelial damage in cardiac ischemia/reperfusion (I/R) injury, sepsis, and diabetes. It delves into the intricate signaling pathways involved in endothelial cell injury, emphasizing the roles of oxidative stress, mitochondrial dysfunction, inflammatory mediators, and barrier damage. Current treatment strategies-ranging from pharmacological interventions to regenerative approaches and lifestyle modifications-face ongoing challenges and limitations. Overall, this review highlights the importance of understanding endothelial cell injury within the context of various diseases and the necessity for innovative therapeutic methods to improve patient outcomes.
Collapse
Affiliation(s)
- Tian Xia
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Laboratory MedicineMedical School of Chinese PLABeijingChina
| | - Jiachi Yu
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Laboratory MedicineMedical School of Chinese PLABeijingChina
| | - Meng Du
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Clinical LaboratoryHuaian Hospital of Huaian CityHuaianJiangsuChina
| | - Ximeng Chen
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Laboratory MedicineMedical School of Chinese PLABeijingChina
| | - Chengbin Wang
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Laboratory MedicineMedical School of Chinese PLABeijingChina
| | - Ruibing Li
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Laboratory MedicineMedical School of Chinese PLABeijingChina
| |
Collapse
|
9
|
Guan X, Li H, Zhang L, Zhi H. Mechanisms of mitochondrial damage-associated molecular patterns associated with inflammatory response in cardiovascular diseases. Inflamm Res 2025; 74:18. [PMID: 39806203 DOI: 10.1007/s00011-025-01993-x] [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: 10/20/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Cardiovascular diseases (CVDs) continue to be a substantial global healthcare burden despite considerable progress in therapies. The inflammatory response during the progression of CVD has attracted considerable attention. Mitochondria serve as the principal energy source for the heart. In cardiovascular illnesses, mitochondrial homeostasis is disrupted, accompanied by structural and functional impairments. During mitochondrial stress or injury, mitochondrial damage-associated molecular patterns (mtDAMPs), such as mitochondrial DNA, cardiolipin, N-formyl peptide, and adenosine triphosphate, are released to activate pattern recognition receptors and trigger immunological responses. Inflammatory responses mediated by mtDAMPs substantially contribute to the pathophysiology of cardiovascular illnesses. In this review, we discuss the molecular mechanisms by which different mtDAMPs control the inflammatory response, address the pathological consequences of mtDAMPs in inducing or exacerbating the inflammatory response in CVDs, and summarize potential therapeutic targets in relevant experimental studies. Preventing or reducing mtDAMP release may play a role in CVD progression by alleviating the inflammatory response.
Collapse
Affiliation(s)
- Xiuju Guan
- School of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Haitao Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China
| | - Lijuan Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China.
| | - Hongwei Zhi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China.
| |
Collapse
|
10
|
Suzuki Y, Giles WR, Zamponi GW, Kondo R, Imaizumi Y, Yamamura H. Ca 2+ signaling in vascular smooth muscle and endothelial cells in blood vessel remodeling: a review. Inflamm Regen 2024; 44:50. [PMID: 39731196 DOI: 10.1186/s41232-024-00363-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/11/2024] [Indexed: 12/29/2024] Open
Abstract
Vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) act together to regulate blood pressure and systemic blood flow by appropriately adjusting blood vessel diameter in response to biochemical or biomechanical stimuli. Ion channels that are expressed in these cells regulate membrane potential and cytosolic Ca2+ concentration ([Ca2+]cyt) in response to such stimuli. The subsets of these ion channels involved in Ca2+ signaling often form molecular complexes with intracellular molecules via scaffolding proteins. This allows Ca2+ signaling to be tightly controlled in localized areas within the cell, resulting in a balanced vascular tone. When hypertensive stimuli are applied to blood vessels for extended periods, gene expression in these vascular cells can change dramatically. For example, alteration in ion channel expression often induces electrical remodeling that produces a depolarization of the membrane potential and elevated [Ca2+]cyt. Coupled with endothelial dysfunction blood vessels undergo functional remodeling characterized by enhanced vasoconstriction. In addition, pathological challenges to vascular cells can induce inflammatory gene products that may promote leukocyte infiltration, in part through Ca2+-dependent pathways. Macrophages accumulating in the vascular adventitia promote fibrosis through extracellular matrix turnover, and cause structural remodeling of blood vessels. This functional and structural remodeling often leads to chronic hypertension affecting not only blood vessels, but also multiple organs including the brain, kidneys, and heart, thus increasing the risk of severe cardiovascular events. In this review, we outline recent advances in multidisciplinary research concerning Ca2+ signaling in VSMCs and ECs, with an emphasis on the mechanisms underlying functional and structural vascular remodeling.
Collapse
Affiliation(s)
- Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.
| | - Wayne R Giles
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Rubii Kondo
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Yuji Imaizumi
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| |
Collapse
|
11
|
Chen X, Yu L, Meng S, Zhao J, Huang X, Wang Z, Zhou Z, Huang Y, Hong T, Duan J, Su T, Cao Z, Chi Y, Huang T, Wang H. Inhibition of TREM-1 ameliorates angiotensin II-induced atrial fibrillation by attenuating macrophage infiltration and inflammation through the PI3K/AKT/FoxO3a signaling pathway. Cell Signal 2024; 124:111458. [PMID: 39384003 DOI: 10.1016/j.cellsig.2024.111458] [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: 07/31/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
Inflammation and infiltration of immune cells are intricately linked to the pathogenesis of atrial fibrillation (AF). Triggering receptor expressed on myeloid cells-1 (TREM-1), an enhancer of inflammation, is implicated in various cardiovascular disorders. However, the precise role and potential mechanisms of TREM-1 in the development of AF remain ambiguous. Atrial samples from patients with AF were used to assess the expression levels of TREM-1. An angiotensin II (Ang II)-induced AF mouse model was established to assess the functionality of TREM-1. Cardiac function and AF inducibility were assessed through echocardiography, programmed transvenous cardiac pacing, and atrial electrophysiological mapping. Peripheral blood and atrial inflammatory cells were assessed using flow cytometry. Using histology, bulk RNA sequencing, biochemical analyses, and cell cultures, the mechanistic role of TREM-1 in AF was elucidated. TREM-1 expression was upregulated and co-localized with macrophages in the atria of patients with AF. Pharmacological inhibition of TREM-1 decreased Ang II-induced atrial enlargement and electrical remodeling. TREM-1 inhibition also ameliorated Ang II-induced NLRP3 inflammasome activation, inflammatory factor release, atrial fibrosis, and macrophage infiltration. Transcriptomic analysis revealed that TREM-1 modulates Ang II-induced inflammation through the PI3K/AKT/FoxO3a signaling pathway. In vitro studies further supported these findings, demonstrating that TREM-1 activation exacerbates Ang II-induced inflammation, while overexpression of FoxO3a counteracts this effect. This study discovered the critical role of TREM-1 in the pathogenesis of AF and its underlying molecular mechanisms. Inhibition of TREM-1 provides a new therapeutic strategy for the treatment of AF.
Collapse
Affiliation(s)
- Xin Chen
- Postgraduate Training Base of General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Liming Yu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Shan Meng
- Postgraduate Training Base of General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Jikai Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Xinyi Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Zhishang Wang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Zijun Zhou
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Yuting Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Tao Hong
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Postgraduate College, Dalian Medical University, Dalian, Liaoning 116000, PR China; Pediatric Surgery Ward, Fuwai Hospital Chinese Academy of Medical Sciences, ShenZhen 518000, PR China
| | - Jinfeng Duan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Postgraduate College, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Tong Su
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110167, PR China
| | - Zijun Cao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Postgraduate College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, PR China
| | - Yanbang Chi
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Department of Obstetrics and Gynecology, General Hospital of Northern Theater Command, Shenyang 110016, PR China
| | - Tao Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| | - Huishan Wang
- Postgraduate Training Base of General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
12
|
Nagata D, Hishida E. Elucidating the complex interplay between chronic kidney disease and hypertension. Hypertens Res 2024; 47:3409-3422. [PMID: 39415028 DOI: 10.1038/s41440-024-01937-8] [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: 05/12/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/18/2024]
Abstract
Chronic kidney disease (CKD) and hypertension share a complex relationship, each exacerbating the progression of the other. CKD contributes to hypertension by decreasing renal function, leading to fluid retention and increased plasma volume, whereas hypertension exacerbates CKD by increasing glomerular pressure and causing renal damage. This review examines the intertwined nature of CKD and hypertension, exploring the factors driving hypertension in CKD and how hypertension accelerates CKD progression. It discusses the role of the renin-angiotensin system and inflammatory cytokines in this relationship, as well as the potential of blood pressure management to slow renal decline. While studies suggest that meticulous blood pressure control can help attenuate CKD progression, optimal management strategies remain unclear and require further investigation. This review also evaluates the evidence surrounding strict antihypertensive therapy in patients with CKD, considering both diabetic and non-diabetic cases. It recommends blood pressure targets based on CKD stage and presence of diabetes, emphasizing the importance of individualized treatment approaches. Renin-angiotensin system inhibitors are highlighted as a key pharmacological intervention due to their renal protective effects, particularly in patients with CKD with proteinuria. However, evidence regarding their efficacy in patients with CKD but without proteinuria is inconclusive. This review underscores the need for comprehensive approaches to effectively address the intertwined nature of CKD and hypertension and calls for further research to optimize clinical management strategies in this complex interplay.
Collapse
Affiliation(s)
- Daisuke Nagata
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Departments of Internal Medicine, Division of Nephrology, Tochigi, Japan.
| | - Erika Hishida
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Departments of Internal Medicine, Division of Nephrology, Tochigi, Japan.
| |
Collapse
|
13
|
R Muralitharan R, Marques FZ, O'Donnell JA. Recent advancements in targeting the immune system to treat hypertension. Eur J Pharmacol 2024; 983:177008. [PMID: 39304109 DOI: 10.1016/j.ejphar.2024.177008] [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: 05/15/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Hypertension is the key leading risk factor for death globally, affecting ∼1.3 billion adults, particularly in low- and middle-income countries. Most people living with hypertension have uncontrolled high blood pressure, increasing their likelihood of cardiovascular events. Significant issues preventing blood pressure control include lack of diagnosis, treatment, and response to existing therapy. For example, monotherapy and combination therapy are often unable to lower blood pressure to target levels. New therapies are urgently required to tackle this issue, particularly those that target the mechanisms behind hypertension instead of treating its symptoms. Acting via an increase in systemic and tissue-specific inflammation, the immune system is a critical contributor to blood pressure regulation and is considered an early mechanism leading to hypertension development. Here, we review the immune system's role in hypertension, evaluate clinical trials that target inflammation, and discuss knowledge gaps in pre-clinical and clinical data. We examine the effects of anti-inflammatory drugs colchicine and methotrexate on hypertension and evaluate the blockade of pro-inflammatory cytokines IL-1β and TNF-α on blood pressure in clinical trials. Lastly, we highlight how we can move forward to target specific components of the immune system to lower blood pressure. This includes targeting isolevuglandins, which accumulate in dendritic cells to promote T cell activation and cytokine production in salt-induced hypertension. We discuss the potential of the dietary fibre-derived metabolites short-chain fatty acids, which have anti-inflammatory and blood pressure-lowering effects via the gut microbiome. This would limit adverse events, leading to improved medication adherence and better blood pressure control.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, VIC, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, VIC, Australia; Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Seale B, Slotabec L, Nguyen JD, Wang H, Patterson C, Filho F, Rouhi N, Adenawoola MI, Li J. Sestrin2 serves as a scaffold protein to maintain cardiac energy and metabolic homeostasis during pathological stress. FASEB J 2024; 38:e70106. [PMID: 39404019 PMCID: PMC11698584 DOI: 10.1096/fj.202401404r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024]
Abstract
Cardiovascular diseases (CVDs) are a leading cause of morbidity and mortality worldwide. Metabolic imbalances and pathological stress often contribute to increased mortality. Sestrin2 (Sesn2) is a stress-inducible protein crucial in maintaining cardiac energy and metabolic homeostasis under pathological conditions. Sesn2 is upregulated in response to various stressors, including oxidative stress, hypoxia, and energy depletion, and mediates multiple cellular pathways to enhance antioxidant defenses, promote autophagy, and inhibit inflammation. This review explores the mechanisms through which Sesn2 regulates these pathways, focusing on the AMPK-mTORC1, Sesn2-Nrf2, and HIF1α-Sesn2 pathways, among others. We can identify the potential therapeutic targets for treating CVDs and related metabolic disorders by comprehending these complex mechanisms. Sesn2's unique ability to respond thoroughly to metabolic challenges, oxidative stress, and inflammation makes it a promising prospect for enhancing cardiac health and resilience against pathological stress.
Collapse
Affiliation(s)
- Blaise Seale
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Lily Slotabec
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Research, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Jennie D. Nguyen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Hao Wang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Cory Patterson
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Fernanda Filho
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael I. Adenawoola
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Research, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
15
|
Chen PH, Chang CK, Lin YK, Chiang SJ, Trang NN. Association of circulating monocyte number and monocyte-lymphocyte ratio with cardiovascular disease in patients with bipolar disorder. BMC Psychiatry 2024; 24:679. [PMID: 39394117 PMCID: PMC11468788 DOI: 10.1186/s12888-024-06105-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 09/20/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) is the leading cause of excessive and premature mortality in patients with bipolar disorder (BD). Despite immune cells participating considerably in the pathogenesis of CVD, limited data are available regarding leukocyte phenotypes in patients with BD and CVD. This study aimed to evaluate associations between circulating leukocyte subset and CVD among patients with BD. METHODS A total of 109 patients with BD-I and cardiologist-confirmed CVD diagnosis (i.e., case) were matched with 109 BD-I patients without CVD (i.e., control) according to the age (± 2 years), sex, and date of most recent psychiatric admission because of acute mood episode (± 2 years). Leukocyte subset data were retrieved from complete blood count tests performed on the next morning after the most recent acute psychiatric admission. RESULTS During the most recent acute psychiatric hospitalization, circulating monocyte counts in the case group were significantly higher than those in the age- and sex-matched controls (p = 0.020). In addition, monocyte-lymphocyte ratios (MLRs) in the case group were significantly higher than those in the control group (p = 0.032). Multiple logistic regression showed that together with serum levels of uric acid and manic symptoms, circulating monocyte counts (95% CI, OR: 1.01-1.05) and MLRs (95% CI, OR: 1.01-1.09) were significantly associated with CVD in patients with BD, respectively. CONCLUSIONS Monocyte activation in an acute manic episode may play a critical role in the pathogenesis of CVD among patients with BD. Future research is required to investigate markers of monocyte activation and indices of cardiovascular structure and function across the different mood states of BD.
Collapse
Affiliation(s)
- Pao-Huan Chen
- Department of Psychiatry, Taipei Medical University Hospital, Taipei, Taiwan.
- Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Chi-Kang Chang
- Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan
| | - Yen-Kuang Lin
- Graduate Institute of Athletics and Coaching Science, National Taiwan Sport University, Taoyuan, Taiwan
| | - Shuo-Ju Chiang
- Division of Cardiology, Department of Internal Medicine, Taipei City Hospital Yangming Branch, Taipei, Taiwan
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | | |
Collapse
|
16
|
Polizio AH, Marino L, Duk-Min K, Yura Y, Rolauer L, Cochran JD, Evans MA, Park E, Doviak H, Miura-Yura E, Good ME, Wolpe AG, Grandoch M, Isakson B, Walsh K. Experimental TET2 Clonal Hematopoiesis Predisposes to Renal Hypertension Through an Inflammasome-Mediated Mechanism. Circ Res 2024; 135:933-950. [PMID: 39234670 PMCID: PMC11519839 DOI: 10.1161/circresaha.124.324492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Hypertension incidence increases with age and represents one of the most prevalent risk factors for cardiovascular disease. Clonal events in the hematopoietic system resulting from somatic mutations in driver genes are prevalent in elderly individuals who lack overt hematologic disorders. This condition is referred to as age-related clonal hematopoiesis (CH), and it is a newly recognized risk factor for cardiovascular disease. It is not known whether CH and hypertension in the elderly are causally related and, if so, what are the mechanistic features. METHODS A murine model of adoptive bone marrow transplantation was employed to examine the interplay between Tet2 (ten-eleven translocation methylcytosine dioxygenase 2) clonal hematopoiesis and hypertension. RESULTS In this model, a subpressor dose of Ang II (angiotensin II) resulted in elevated systolic and diastolic blood pressure as early as 1 day after challenge. These conditions led to the expansion of Tet2-deficient proinflammatory monocytes and bone marrow progenitor populations. Tet2 deficiency promoted renal CCL5 (C-C motif ligand 5) chemokine expression and macrophage infiltration into the kidney. Consistent with macrophage involvement, Tet2 deficiency in myeloid cells promoted hypertension when mice were treated with a subpressor dose of Ang II. The hematopoietic Tet2-/- condition led to sodium retention, renal inflammasome activation, and elevated levels of IL (interleukin)-1β and IL-18. Analysis of the sodium transporters indicated NCC (sodium-chloride symporter) and NKCC2 (Na+-K+-Cl- cotransporter 2) activation at residues Thr53 and Ser105, respectively. Administration of the NLRP3 (NLR family pyrin domain containing 3) inflammasome inhibitor MCC950 reversed the hypertensive state, sodium retention, and renal transporter activation. CONCLUSIONS Tet2-mediated CH sensitizes mice to a hypertensive stimulus. Mechanistically, the expansion of hematopoietic Tet2-deficient cells promotes hypertension due to elevated renal immune cell infiltration and activation of the NLRP3 inflammasome, with consequences on sodium retention. These data indicate that carriers of TET2 CH could be at elevated risk for the development of hypertension and that immune modulators could be useful in treating hypertension in this patient population.
Collapse
Affiliation(s)
- Ariel H. Polizio
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Lucila Marino
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kyung Duk-Min
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Yoshimitsu Yura
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Luca Rolauer
- Institute of Translational Pharmacology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jesse D. Cochran
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Megan A. Evans
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Eunbee Park
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Heather Doviak
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Emiri Miura-Yura
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Miranda E. Good
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA, 02111, USA
| | | | - Maria Grandoch
- Institute of Translational Pharmacology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | - Brant Isakson
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kenneth Walsh
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
17
|
Roy S, Srinivasan VR, Arunagiri S, Mishra N, Bhatia A, Shejale KP, Prajapati KP, Kar K, Anand BG. Molecular insights into the phase transition of lysozyme into amyloid nanostructures: Implications of therapeutic strategies in diverse pathological conditions. Adv Colloid Interface Sci 2024; 331:103205. [PMID: 38875805 DOI: 10.1016/j.cis.2024.103205] [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: 09/17/2023] [Revised: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 06/16/2024]
Abstract
Lysozyme, a well-known bacteriolytic enzyme, exhibits a fascinating yet complex behavior when it comes to protein aggregation. Under certain conditions, this enzyme undergoes flexible transformation, transitioning from partially unfolded intermediate units of native conformers into complex cross-β-rich nano fibrillar amyloid architectures. Formation of such lysozyme amyloids has been implicated in a multitude of pathological and medical severities, like hepatic dysfunction, hepatomegaly, splenic rupture as well as spleen dysfunction, nephropathy, sicca syndrome, renal dysfunction, renal amyloidosis, and systemic amyloidosis. In this comprehensive review, we have attempted to provide in-depth insights into the aggregating behavior of lysozyme across a spectrum of variables, including concentrations, temperatures, pH levels, and mutations. Our objective is to elucidate the underlying mechanisms that govern lysozyme's aggregation process and to unravel the complex interplay between its structural attributes. Moreover, this work has critically examined the latest advancements in the field, focusing specifically on novel strategies and systems, that have been implemented to delay or inhibit the lysozyme amyloidogenesis. Apart from this, we have tried to explore and advance our fundamental understanding of the complex processes involved in lysozyme aggregation. This will help the research community to lay a robust foundation for screening, designing, and formulating targeted anti-amyloid therapeutics offering improved treatment modalities and interventions not only for lysozyme-linked amyloidopathy but for a wide range of amyloid-related disorders.
Collapse
Affiliation(s)
- Sindhujit Roy
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Venkat Ramanan Srinivasan
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Subash Arunagiri
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Nishant Mishra
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Anubhuti Bhatia
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Kiran P Shejale
- Dept. of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, South Korea
| | - Kailash Prasad Prajapati
- Biophysical and Biomaterials Research Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Karunakar Kar
- Biophysical and Biomaterials Research Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India..
| | - Bibin Gnanadhason Anand
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India..
| |
Collapse
|
18
|
Liao Y, Zhu L. At the heart of inflammation: Unravelling cardiac resident macrophage biology. J Cell Mol Med 2024; 28:e70050. [PMID: 39223947 PMCID: PMC11369210 DOI: 10.1111/jcmm.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular disease remains one of the leading causes of death globally. Recent advancements in sequencing technologies have led to the identification of a unique population of macrophages within the heart, termed cardiac resident macrophages (CRMs), which exhibit self-renewal capabilities and play crucial roles in regulating cardiac homeostasis, inflammation, as well as injury and repair processes. This literature review aims to elucidate the origin and phenotypic characteristics of CRMs, comprehensively outline their contributions to cardiac homeostasis and further summarize their functional roles and molecular mechanisms implicated in the onset and progression of cardiovascular diseases. These insights are poised to pave the way for novel therapeutic strategies centred on targeted interventions based on the distinctive properties of resident macrophages.
Collapse
Affiliation(s)
- Yingnan Liao
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
- Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduSichuanChina
| | - Liyuan Zhu
- Center of Clinical Pharmacology, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
19
|
Nguyen BA, Alexander MR, Harrison DG. Immune mechanisms in the pathophysiology of hypertension. Nat Rev Nephrol 2024; 20:530-540. [PMID: 38658669 PMCID: PMC12060254 DOI: 10.1038/s41581-024-00838-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
Hypertension is a leading risk factor for morbidity and mortality worldwide. Despite current anti-hypertensive therapies, most individuals with hypertension fail to achieve adequate blood pressure control. Moreover, even with adequate control, a residual risk of cardiovascular events and associated organ damage remains. These findings suggest that current treatment modalities are not addressing a key element of the underlying pathology. Emerging evidence implicates immune cells as key mediators in the development and progression of hypertension. In this Review, we discuss our current understanding of the diverse roles of innate and adaptive immune cells in hypertension, highlighting key findings from human and rodent studies. We explore mechanisms by which these immune cells promote hypertensive pathophysiology, shedding light on their multifaceted involvement. In addition, we highlight advances in our understanding of autoimmunity, HIV and immune checkpoints that provide valuable insight into mechanisms of chronic and dysregulated inflammation in hypertension.
Collapse
Affiliation(s)
- Bianca A Nguyen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Matthew R Alexander
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, USA
| | - David G Harrison
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, USA.
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
20
|
Harrison DG, Patrick DM. Immune Mechanisms in Hypertension. Hypertension 2024; 81:1659-1674. [PMID: 38881474 PMCID: PMC11254551 DOI: 10.1161/hypertensionaha.124.21355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
It is now apparent that immune mediators including complement, cytokines, and cells of the innate and adaptive immune system contribute not only to blood pressure elevation but also to the target organ damage that occurs in response to stimuli like high salt, aldosterone, angiotensin II, and sympathetic outflow. Alterations of vascular hemodynamic factors, including microvascular pulsatility and shear forces, lead to vascular release of mediators that affect myeloid cells to become potent antigen-presenting cells and promote T-cell activation. Research in the past 2 decades has defined specific biochemical and molecular pathways that are engaged by these stimuli and an emerging paradigm is these not only lead to immune activation, but that products of immune cells, including cytokines, reactive oxygen species, and metalloproteinases act on target cells to further raise blood pressure in a feed-forward fashion. In this review, we will discuss these molecular and pathophysiological events and discuss clinical interventions that might prove effective in quelling this inflammatory process in hypertension and related cardiovascular diseases.
Collapse
Affiliation(s)
- David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - David M. Patrick
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Veterans Affairs, Nashville, TN 37212
| |
Collapse
|
21
|
Strohm L, Daiber A, Ubbens H, Krishnankutty R, Oelze M, Kuntic M, Hahad O, Klein V, Hoefer IE, von Kriegsheim A, Kleinert H, Atzler D, Lurz P, Weber C, Wild PS, Münzel T, Knosalla C, Lutgens E, Daub S. Role of inflammatory signaling pathways involving the CD40-CD40L-TRAF cascade in diabetes and hypertension-insights from animal and human studies. Basic Res Cardiol 2024; 119:1-18. [PMID: 38554187 PMCID: PMC11319409 DOI: 10.1007/s00395-024-01045-1] [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: 11/28/2023] [Revised: 02/09/2024] [Accepted: 03/02/2024] [Indexed: 04/01/2024]
Abstract
CD40L-CD40-TRAF signaling plays a role in atherosclerosis progression and affects the pathogenesis of coronary heart disease (CHD). We tested the hypothesis that CD40L-CD40-TRAF signaling is a potential therapeutic target in hyperlipidemia, diabetes, and hypertension. In mouse models of hyperlipidemia plus diabetes (db/db mice) or hypertension (1 mg/kg/d angiotensin-II for 7 days), TRAF6 inhibitor treatment (2.5 mg/kg/d for 7 or 14 days) normalized markers of oxidative stress and inflammation. As diabetes and hypertension are important comorbidities aggravating CHD, we explored whether the CD40L-CD40-TRAF signaling cascade and their associated inflammatory pathways are expressed in CHD patients suffering from comorbidities. Therefore, we analyzed vascular bypass material (aorta or internal mammary artery) and plasma from patients with CHD with diabetes and/or hypertension. Our Olink targeted plasma proteomic analysis using the IMMUNO-ONCOLOGY panel revealed a pattern of step-wise increase for 13/92 markers of low-grade inflammation with significant changes. CD40L or CD40 significantly correlated with 38 or 56 other inflammatory targets. In addition, specific gene clusters that correlate with the comorbidities were identified in isolated aortic mRNA of CHD patients through RNA-sequencing. These signaling clusters comprised CD40L-CD40-TRAF, immune system, hemostasis, muscle contraction, metabolism of lipids, developmental biology, and apoptosis. Finally, immunological analysis revealed key markers correlated with comorbidities in CHD patients, such as CD40L, NOX2, CD68, and 3-nitrotyrosine. These data indicate that comorbidities increase inflammatory pathways in CHD, and targeting these pathways will be beneficial in reducing cardiovascular events in CHD patients with comorbidities.
Collapse
Affiliation(s)
- Lea Strohm
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany.
- Universitätsmedizin der Johannes Gutenberg-Universität Zentrum für Kardiologie 1, Labor für Molekulare Kardiologie, Geb. 605, Raum 3.262, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Henning Ubbens
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | | | - Matthias Oelze
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Marin Kuntic
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Omar Hahad
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany
| | - Veronique Klein
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Imo E Hoefer
- Central Diagnostic Laboratory, UMC Utrecht, Utrecht, The Netherlands
| | | | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Philipp Lurz
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Philipp S Wild
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
- Clinical Epidemiology and Systems Medicine, Center for Thrombosis and Hemostasis, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
- Systems Medicine, Institute of Molecular Biology (IMB), Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany
| | - Christoph Knosalla
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, USA
| | - Steffen Daub
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
22
|
Kuntic M, Hahad O, Al-Kindi S, Oelze M, Lelieveld J, Daiber A, Münzel T. Pathomechanistic Synergy Between Particulate Matter and Traffic Noise-Induced Cardiovascular Damage and the Classical Risk Factor Hypertension. Antioxid Redox Signal 2024. [PMID: 38874533 DOI: 10.1089/ars.2024.0659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Affiliation(s)
- Marin Kuntic
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| | - Omar Hahad
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| | - Sadeer Al-Kindi
- Cardiovascular Prevention & Wellness and Center for CV Computational & Precision Health, Houston Methodist DeBakey Heart & Vascular Center, Houston, Texas, USA
| | - Matthias Oelze
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jos Lelieveld
- Max Planck Institute for Chemistry, Atmospheric Chemistry, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| |
Collapse
|
23
|
Totoń-Żurańska J, Mikolajczyk TP, Saju B, Guzik TJ. Vascular remodelling in cardiovascular diseases: hypertension, oxidation, and inflammation. Clin Sci (Lond) 2024; 138:817-850. [PMID: 38920058 DOI: 10.1042/cs20220797] [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: 09/26/2023] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024]
Abstract
Optimal vascular structure and function are essential for maintaining the physiological functions of the cardiovascular system. Vascular remodelling involves changes in vessel structure, including its size, shape, cellular and molecular composition. These changes result from multiple risk factors and may be compensatory adaptations to sustain blood vessel function. They occur in diverse cardiovascular pathologies, from hypertension to heart failure and atherosclerosis. Dynamic changes in the endothelium, fibroblasts, smooth muscle cells, pericytes or other vascular wall cells underlie remodelling. In addition, immune cells, including macrophages and lymphocytes, may infiltrate vessels and initiate inflammatory signalling. They contribute to a dynamic interplay between cell proliferation, apoptosis, migration, inflammation, and extracellular matrix reorganisation, all critical mechanisms of vascular remodelling. Molecular pathways underlying these processes include growth factors (e.g., vascular endothelial growth factor and platelet-derived growth factor), inflammatory cytokines (e.g., interleukin-1β and tumour necrosis factor-α), reactive oxygen species, and signalling pathways, such as Rho/ROCK, MAPK, and TGF-β/Smad, related to nitric oxide and superoxide biology. MicroRNAs and long noncoding RNAs are crucial epigenetic regulators of gene expression in vascular remodelling. We evaluate these pathways for potential therapeutic targeting from a clinical translational perspective. In summary, vascular remodelling, a coordinated modification of vascular structure and function, is crucial in cardiovascular disease pathology.
Collapse
Affiliation(s)
- Justyna Totoń-Żurańska
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz P Mikolajczyk
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Blessy Saju
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, U.K
| | - Tomasz J Guzik
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, U.K
| |
Collapse
|
24
|
Peter JK, Umene R, Wu CH, Nakamura Y, Washimine N, Yamamoto R, Ngugi C, Linge K, Kweri JK, Inoue T. Renal macrophages induce hypertension and kidney fibrosis in Angiotensin II salt mice model. Biochem Biophys Res Commun 2024; 715:149997. [PMID: 38678782 DOI: 10.1016/j.bbrc.2024.149997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/12/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
The immune system is involved in hypertension development with different immune cells reported to have either pro or anti-hypertensive effects. In hypertension, immune cells have been thought to infiltrate blood pressure-regulating organs, resulting in either elevation or reduction of blood pressure. There is controversy over whether macrophages play a detrimental or beneficial role in the development of hypertension, and the few existing studies have yielded conflicting results. This study aimed to determine the effects of angiotensin II (Ang II) salt-induced hypertension on renal immune cells and to determine whether renal macrophages are involved in the induction of hypertension. Hypertension was induced by administration of Ang II and saline for two weeks. The effects of hypertension on kidney immune cells were assessed using flow cytometry. Macrophage infiltration in the kidney was assessed by immunohistochemistry and kidney fibrosis was assessed using trichrome stain and kidney real time-qPCR. Liposome encapsulated clodronate was used to deplete macrophages in C57BL/6J mice and investigate the direct role of macrophages in hypertension induction. Ang II saline mice group developed hypertension, had increased renal macrophages, and had increased expression of Acta2 and Col1a1 and kidney fibrotic areas. Macrophage depletion blunted hypertension development and reduced the expression of Acta2 and Col1a1 in the kidney and kidney fibrotic areas in Ang II saline group. The results of this study demonstrate that macrophages infiltrate the kidneys and increase kidney fibrosis in Ang II salt-induced hypertension, and depletion of macrophages suppresses the development of hypertension and decreases kidney fibrosis. This indicates that macrophages play a direct role in hypertension development. Hence macrophages have a potential to be considered as therapeutic target in hypertension management.
Collapse
Affiliation(s)
- Joseph Kasyoki Peter
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan; Department of Medical Physiology, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya; Department of Clinical Medicine, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Ryusuke Umene
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan; Department of Nephrology, Graduate School of Biomedical Sciences, Nagasaki University, Japan.
| | - Chia-Hsien Wu
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Yasuna Nakamura
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Norito Washimine
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan; Department of Nephrology, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Ryoko Yamamoto
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Caroline Ngugi
- Department of Medical Microbiology, School of Biomedical Sciences, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Kavoo Linge
- Department of Medical Physiology, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Joseph K Kweri
- Department of Human Anatomy, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Tsuyoshi Inoue
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan.
| |
Collapse
|
25
|
Grishanova AY, Perepechaeva ML. Kynurenic Acid/AhR Signaling at the Junction of Inflammation and Cardiovascular Diseases. Int J Mol Sci 2024; 25:6933. [PMID: 39000041 PMCID: PMC11240928 DOI: 10.3390/ijms25136933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Persistent systemic chronic inflammatory conditions are linked with many pathologies, including cardiovascular diseases (CVDs), a leading cause of death across the globe. Among various risk factors, one of the new possible contributors to CVDs is the metabolism of essential amino acid tryptophan. Proinflammatory signals promote tryptophan metabolism via the kynurenine (KYN) pathway (KP), thereby resulting in the biosynthesis of several immunomodulatory metabolites whose biological effects are associated with the development of symptoms and progression of various inflammatory diseases. Some participants in the KP are agonists of aryl hydrocarbon receptor (AhR), a central player in a signaling pathway that, along with a regulatory influence on the metabolism of environmental xenobiotics, performs a key immunomodulatory function by triggering various cellular mechanisms with the participation of endogenous ligands to alleviate inflammation. An AhR ligand with moderate affinity is the central metabolite of the KP: KYN; one of the subsequent metabolites of KYN-kynurenic acid (KYNA)-is a more potent ligand of AhR. Understanding the role of AhR pathway-related metabolites of the KP that regulate inflammatory factors in cells of the cardiovascular system is interesting and important for achieving effective treatment of CVDs. The purpose of this review was to summarize the results of studies about the participation of the KP metabolite-KYNA-and of the AhR signaling pathway in the regulation of inflammation in pathological conditions of the heart and blood vessels and about the possible interaction of KYNA with AhR signaling in some CVDs.
Collapse
Affiliation(s)
| | - Maria L. Perepechaeva
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2, Novosibirsk 630060, Russia;
| |
Collapse
|
26
|
Li CX, Yue L. The Multifaceted Nature of Macrophages in Cardiovascular Disease. Biomedicines 2024; 12:1317. [PMID: 38927523 PMCID: PMC11201197 DOI: 10.3390/biomedicines12061317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/01/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
As the leading cause of mortality worldwide, cardiovascular disease (CVD) represents a variety of heart diseases and vascular disorders, including atherosclerosis, aneurysm, ischemic injury in the heart and brain, arrythmias, and heart failure. Macrophages, a diverse population of immune cells that can promote or suppress inflammation, have been increasingly recognized as a key regulator in various processes in both healthy and disease states. In healthy conditions, these cells promote the proper clearance of cellular debris, dead and dying cells, and provide a strong innate immune barrier to foreign pathogens. However, macrophages can play a detrimental role in the progression of disease as well, particularly those inflammatory in nature. This review will focus on the current knowledge regarding the role of macrophages in cardiovascular diseases.
Collapse
Affiliation(s)
- Cindy X. Li
- Department of Cell Biology, Pat and Jim Calhoun Cardiovascular Center, University of Connecticut Health Center, Farmington, CT 06030, USA;
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Lixia Yue
- Department of Cell Biology, Pat and Jim Calhoun Cardiovascular Center, University of Connecticut Health Center, Farmington, CT 06030, USA;
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
27
|
Balhara M, Neikirk K, Marshall A, Hinton A, Kirabo A. Endoplasmic Reticulum Stress in Hypertension and Salt Sensitivity of Blood Pressure. Curr Hypertens Rep 2024; 26:273-290. [PMID: 38602583 PMCID: PMC11166838 DOI: 10.1007/s11906-024-01300-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2024] [Indexed: 04/12/2024]
Abstract
PURPOSE OF REVIEW Hypertension is a principal risk factor for cardiovascular morbidity and mortality, with its severity exacerbated by high sodium intake, particularly in individuals with salt-sensitive blood pressure. However, the mechanisms underlying hypertension and salt sensitivity are only partly understood. Herein, we review potential interactions in hypertension pathophysiology involving the immune system, endoplasmic reticulum (ER) stress, the unfolded protein response (UPR), and proteostasis pathways; identify knowledge gaps; and discuss future directions. RECENT FINDINGS Recent advancements by our research group and others reveal interactions within and between adaptive and innate immune responses in hypertension pathophysiology. The salt-immune-hypertension axis is further supported by the discovery of the role of dendritic cells in hypertension, marked by isolevuglandin (IsoLG) formation. Alongside these broadened understandings of immune-mediated salt sensitivity, the contributions of T cells to hypertension have been recently challenged by groups whose findings did not support increased resistance of Rag-1-deficient mice to Ang II infusion. Hypertension has also been linked to ER stress and the UPR. Notably, a holistic approach is needed because the UPR engages in crosstalk with autophagy, the ubiquitin proteasome, and other proteostasis pathways, that may all involve hypertension. There is a critical need for studies to establish cause and effect relationships between ER stress and the UPR in hypertension pathophysiology in humans and to determine whether the immune system and ER stress function mainly to exacerbate or initiate hypertension and target organ injury. This review of recent studies proposes new avenues for future research for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Maria Balhara
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Andrea Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA.
- Vanderbilt Center for Immunobiology, Nashville, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, USA.
- Vanderbilt Institute for Global Health, Nashville, USA.
| |
Collapse
|
28
|
Guzik TJ, Nosalski R, Maffia P, Drummond GR. Immune and inflammatory mechanisms in hypertension. Nat Rev Cardiol 2024; 21:396-416. [PMID: 38172242 DOI: 10.1038/s41569-023-00964-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 01/05/2024]
Abstract
Hypertension is a global health problem, with >1.3 billion individuals with high blood pressure worldwide. In this Review, we present an inflammatory paradigm for hypertension, emphasizing the crucial roles of immune cells, cytokines and chemokines in disease initiation and progression. T cells, monocytes, macrophages, dendritic cells, B cells and natural killer cells are all implicated in hypertension. Neoantigens, the NLRP3 inflammasome and increased sympathetic outflow, as well as cytokines (including IL-6, IL-7, IL-15, IL-18 and IL-21) and a high-salt environment, can contribute to immune activation in hypertension. The activated immune cells migrate to target organs such as arteries (especially the perivascular fat and adventitia), kidneys, the heart and the brain, where they release effector cytokines that elevate blood pressure and cause vascular remodelling, renal damage, cardiac hypertrophy, cognitive impairment and dementia. IL-17 secreted by CD4+ T helper 17 cells and γδ T cells, and interferon-γ and tumour necrosis factor secreted by immunosenescent CD8+ T cells, exert crucial effector roles in hypertension, whereas IL-10 and regulatory T cells are protective. Effector mediators impair nitric oxide bioavailability, leading to endothelial dysfunction and increased vascular contractility. Inflammatory effector mediators also alter renal sodium and water balance and promote renal fibrosis. These mechanisms link hypertension with obesity, autoimmunity, periodontitis and COVID-19. A comprehensive understanding of the immune and inflammatory mechanisms of hypertension is crucial for safely and effectively translating the findings to clinical practice.
Collapse
Affiliation(s)
- Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK.
- Department of Medicine and Omicron Medical Genomics Laboratory, Jagiellonian University, Collegium Medicum, Kraków, Poland.
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK.
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Pasquale Maffia
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Grant R Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
29
|
Zhang X, Li G, Wu W, Li B. Causal role of immune cells in hypertension: a bidirectional Mendelian randomization study. Front Cardiovasc Med 2024; 11:1375704. [PMID: 38859818 PMCID: PMC11163045 DOI: 10.3389/fcvm.2024.1375704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/10/2024] [Indexed: 06/12/2024] Open
Abstract
Background Although Hypertension (HTN) is considered to be a cardiovascular disease caused by multiple factors, the cause of it is still unknown. In this study, we aim to find out whether circulating immune cell characteristics have an impact on susceptibility to HTN. Methods This study employed a comprehensive two-sample Mendelian randomization (MR) analysis to investigate the causal association between immune cell characteristics and HTN. Utilizing publicly accessible genetic data, we examined the causal relationship between HTN and the susceptibility to 731 immune cell signatures. To ensure the reliability and validity of the findings, a comprehensive sensitivity analysis was conducted to assess heterogeneity, confirm the robustness of the results and evaluate the presence of horizontal pleiotropy. Results After FDR correction, immune phenotype had an effect on HTN. In our study, one immunophenotype was identified as being positively associated with HTN risk significance: HLA DR on CD33- HLA DR+. In addition, we examined 8 immune phenotype with no statistically significant effect of HTN, but it is worth mentioning that they had an unadjusted low P-value phenotype. Conclusions Our MR study by genetic means demonstrated the close relationship between HTN and immune cells, thus providing guidance for future clinical prediction and subsequent treatment of HTN.
Collapse
Affiliation(s)
- Xinhe Zhang
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong First Medical University, Jinan, China
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guanying Li
- Jinan Foreign Language School International Center, Jinan, China
| | - Wei Wu
- Department of Cardiology, Hekou District People Hospital, Dongying, China
| | - Bin Li
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
30
|
Zifkos K, Bochenek ML, Gogiraju R, Robert S, Pedrosa D, Kiouptsi K, Moiko K, Wagner M, Mahfoud F, Poncelet P, Münzel T, Ruf W, Reinhardt C, Panicot-Dubois L, Dubois C, Schäfer K. Endothelial PTP1B Deletion Promotes VWF Exocytosis and Venous Thromboinflammation. Circ Res 2024; 134:e93-e111. [PMID: 38563147 DOI: 10.1161/circresaha.124.324214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Endothelial activation promotes the release of procoagulant extracellular vesicles and inflammatory mediators from specialized storage granules. Endothelial membrane exocytosis is controlled by phosphorylation. We hypothesized that the absence of PTP1B (protein tyrosine phosphatase 1B) in endothelial cells promotes venous thromboinflammation by triggering endothelial membrane fusion and exocytosis. METHODS Mice with inducible endothelial deletion of PTP1B (End.PTP1B-KO) underwent inferior vena cava ligation to induce stenosis and venous thrombosis. Primary endothelial cells from transgenic mice and human umbilical vein endothelial cells were used for mechanistic studies. RESULTS Vascular ultrasound and histology showed significantly larger venous thrombi containing higher numbers of Ly6G (lymphocyte antigen 6 family member G)-positive neutrophils in mice with endothelial PTP1B deletion, and intravital microscopy confirmed the more pronounced neutrophil recruitment following inferior vena cava ligation. RT2 PCR profiler array and immunocytochemistry analysis revealed increased endothelial activation and adhesion molecule expression in primary End.PTP1B-KO endothelial cells, including CD62P (P-selectin) and VWF (von Willebrand factor). Pretreatment with the NF-κB (nuclear factor kappa B) kinase inhibitor BAY11-7082, antibodies neutralizing CD162 (P-selectin glycoprotein ligand-1) or VWF, or arginylglycylaspartic acid integrin-blocking peptides abolished the neutrophil adhesion to End.PTP1B-KO endothelial cells in vitro. Circulating levels of annexin V+ procoagulant endothelial CD62E+ (E-selectin) and neutrophil (Ly6G+) extracellular vesicles were also elevated in End.PTP1B-KO mice after inferior vena cava ligation. Higher plasma MPO (myeloperoxidase) and Cit-H3 (citrullinated histone-3) levels and neutrophil elastase activity indicated neutrophil activation and extracellular trap formation. Infusion of End.PTP1B-KO extracellular vesicles into C57BL/6J wild-type mice most prominently enhanced the recruitment of endogenous neutrophils, and this response was blunted in VWF-deficient mice or by VWF-blocking antibodies. Reduced PTP1B binding and tyrosine dephosphorylation of SNAP23 (synaptosome-associated protein 23) resulting in increased VWF exocytosis and neutrophil adhesion were identified as mechanisms, all of which could be restored by NF-κB kinase inhibition using BAY11-7082. CONCLUSIONS Our findings show that endothelial PTP1B deletion promotes venous thromboinflammation by enhancing SNAP23 phosphorylation, endothelial VWF exocytosis, and neutrophil recruitment.
Collapse
Affiliation(s)
- Konstantinos Zifkos
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
| | - Magdalena L Bochenek
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
- Department of Cardiology, Cardiology I (M.L.B., R.G., K.M., T.M., K.S.), University Medical Center Mainz, Germany
| | - Rajinikanth Gogiraju
- Department of Cardiology, Cardiology I (M.L.B., R.G., K.M., T.M., K.S.), University Medical Center Mainz, Germany
| | - Stéphane Robert
- Aix Marseille University, National Institute of Health and Medical Research (INSERM) 1263, National Research Institute for Agriculture, Food and Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), France (S.R., L.P.-D., C.D.)
| | - Denise Pedrosa
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
| | - Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
| | - Kateryna Moiko
- Department of Cardiology, Cardiology I (M.L.B., R.G., K.M., T.M., K.S.), University Medical Center Mainz, Germany
| | - Mathias Wagner
- Institute of Pathology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany (M.W.)
| | - Felix Mahfoud
- Department of Internal Medicine III, Cardiology, Angiology and Internal Intensive Care Medicine, Saarland University Hospital and Saarland University, Homburg, Germany (F.M.)
| | | | - Thomas Münzel
- Department of Cardiology, Cardiology I (M.L.B., R.G., K.M., T.M., K.S.), University Medical Center Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
| | - Laurence Panicot-Dubois
- Aix Marseille University, National Institute of Health and Medical Research (INSERM) 1263, National Research Institute for Agriculture, Food and Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), France (S.R., L.P.-D., C.D.)
| | - Christophe Dubois
- Aix Marseille University, National Institute of Health and Medical Research (INSERM) 1263, National Research Institute for Agriculture, Food and Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), France (S.R., L.P.-D., C.D.)
| | - Katrin Schäfer
- Department of Cardiology, Cardiology I (M.L.B., R.G., K.M., T.M., K.S.), University Medical Center Mainz, Germany
| |
Collapse
|
31
|
Gan J, Shi Y, Zhao R, Li D, Jin H, Wu M, Liu Z, Li X, Xu A, Li Y, Lin Z, Wu F. Adipose c-Jun NH2-terminal kinase promotes angiotensin II-induced and deoxycorticosterone acetate salt-induced hypertension and vascular dysfunction by inhibition of adiponectin production and activation of SGK1 in mice. J Hypertens 2024; 42:856-872. [PMID: 38164960 DOI: 10.1097/hjh.0000000000003649] [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: 01/03/2024]
Abstract
BACKGROUND Adipose c-Jun NH2-terminal kinase 1/2 (JNK1/2) is a central mediator involved in the development of obesity and its complications. However, the roles of adipose JNK1/2 in hypertension remain elusive. Here we explored the role of adipose JNK1/2 in hypertension. METHODS AND RESULTS The roles of adipose JNK1/2 in hypertension were investigated by evaluating the impact of adipose JNK1/2 inactivation in both angiotensin II (Ang II)-induced and deoxycorticosterone acetate (DOCA) salt-induced hypertensive mice. Specific inactivation of JNK1/2 in adipocytes significantly alleviates Ang II-induced and DOCA salt-induced hypertension and target organ damage in mice. Interestingly, such beneficial effects are also observed in hypertensive mice after oral administration of JNK1/2 inhibitor SP600125. Mechanistically, adipose JNK1/2 acts on adipocytes to reduce the production of adiponectin (APN), then leads to promote serum and glucocorticoid-regulated kinase 1 (SGK1) phosphorylation and increases epithelial Na + channel α-subunit (ENaCα) expression in both renal cells and adipocytes, respectively, finally exacerbates Na + retention. In addition, chronic treatment of recombinant mouse APN significantly augments the beneficial effects of adipose JNK1/2 inactivation in DOCA salt-induced hypertension. By contrast, the blood pressure-lowering effects of adipose JNK1/2 inactivation are abrogated by adenovirus-mediated SGK1 overexpression in Ang II -treated adipose JNK1/2 inactivation mice. CONCLUSION Adipose JNK1/2 promotes hypertension and targets organ impairment via fine-tuning the multiorgan crosstalk among adipose tissue, kidney, and blood vessels.
Collapse
Affiliation(s)
- Jing Gan
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University
| | - Yaru Shi
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
- Department of Pharmacy, the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui
| | - Ruyi Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| | - Dan Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
- Department of clinical pharmacy, the Forth People's Hospital of Liaocheng, Liaocheng
| | - Hua Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| | - Maolan Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| | - Zhen Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong
| | - Yulin Li
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Anzhen Hospital of Capital Medical University, Beijing
| | - Zhuofeng Lin
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University
- The laboratory of Animal Center, Wenzhou Medical University, Wenzhou, China
| | - Fan Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| |
Collapse
|
32
|
Maaliki D, Itani M, Jarrah H, El-Mallah C, Ismail D, El Atie YE, Obeid O, Jaffa MA, Itani HA. Dietary High Salt Intake Exacerbates SGK1-Mediated T Cell Pathogenicity in L-NAME/High Salt-Induced Hypertension. Int J Mol Sci 2024; 25:4402. [PMID: 38673987 PMCID: PMC11050194 DOI: 10.3390/ijms25084402] [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: 03/09/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Sodium chloride (NaCl) activates Th17 and dendritic cells in hypertension by stimulating serum/glucocorticoid kinase 1 (SGK1), a sodium sensor. Memory T cells also play a role in hypertension by infiltrating target organs and releasing proinflammatory cytokines. We tested the hypothesis that the role of T cell SGK1 extends to memory T cells. We employed mice with a T cell deletion of SGK1, SGK1fl/fl × tgCD4cre mice, and used SGK1fl/fl mice as controls. We treated the mice with L-NAME (0.5 mg/mL) for 2 weeks and allowed a 2-week washout interval, followed by a 3-week high-salt (HS) diet (4% NaCl). L-NAME/HS significantly increased blood pressure and memory T cell accumulation in the kidneys and bone marrow of SGK1fl/fl mice compared to knockout mice on L-NAME/HS or groups on a normal diet (ND). SGK1fl/fl mice exhibited increased albuminuria, renal fibrosis, and interferon-γ levels after L-NAME/HS treatment. Myography demonstrated endothelial dysfunction in the mesenteric arterioles of SGK1fl/fl mice. Bone marrow memory T cells were adoptively transferred from either mouse strain after L-NAME/HS administration to recipient CD45.1 mice fed the HS diet for 3 weeks. Only the mice that received cells from SGK1fl/fl donors exhibited increased blood pressure and renal memory T cell infiltration. Our data suggest a new therapeutic target for decreasing hypertension-specific memory T cells and protecting against hypertension.
Collapse
Affiliation(s)
- Dina Maaliki
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (D.M.); (M.I.); (H.J.); (D.I.); (Y.E.E.A.)
| | - Maha Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (D.M.); (M.I.); (H.J.); (D.I.); (Y.E.E.A.)
| | - Hala Jarrah
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (D.M.); (M.I.); (H.J.); (D.I.); (Y.E.E.A.)
| | - Carla El-Mallah
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut 1107, Lebanon; (C.E.-M.); (O.O.)
| | - Diana Ismail
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (D.M.); (M.I.); (H.J.); (D.I.); (Y.E.E.A.)
| | - Yara E. El Atie
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (D.M.); (M.I.); (H.J.); (D.I.); (Y.E.E.A.)
| | - Omar Obeid
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut 1107, Lebanon; (C.E.-M.); (O.O.)
| | - Miran A. Jaffa
- Epidemiology and Population Health Department, Faculty of Health Sciences, American University of Beirut, Beirut 1107, Lebanon;
| | - Hana A. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (D.M.); (M.I.); (H.J.); (D.I.); (Y.E.E.A.)
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
33
|
Ávila-Martínez DV, Mixtega-Ruiz WK, Hurtado-Capetillo JM, Lopez-Franco O, Flores-Muñoz M. Counter-regulatory RAS peptides: new therapy targets for inflammation and fibrotic diseases? Front Pharmacol 2024; 15:1377113. [PMID: 38666016 PMCID: PMC11044688 DOI: 10.3389/fphar.2024.1377113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/18/2024] [Indexed: 04/28/2024] Open
Abstract
The renin-angiotensin system (RAS) is an important cascade of enzymes and peptides that regulates blood pressure, volume, and electrolytes. Within this complex system of reactions, its counter-regulatory axis has attracted attention, which has been associated with the pathophysiology of inflammatory and fibrotic diseases. This review article analyzes the impact of different components of the counter-regulatory axis of the RAS on different pathologies. Of these peptides, Angiotensin-(1-7), angiotensin-(1-9) and alamandine have been evaluated in a wide variety of in vitro and in vivo studies, where not only they counteract the actions of the classical axis, but also exhibit independent anti-inflammatory and fibrotic actions when binding to specific receptors, mainly in heart, kidney, and lung. Other functional peptides are also addressed, which despite no reports associated with inflammation and fibrosis to date were found, they could represent a potential target of study. Furthermore, the association of agonists of the counter-regulatory axis is analyzed, highlighting their contribution to the modulation of the inflammatory response counteracting the development of fibrotic events. This article shows an overview of the importance of the RAS in the resolution of inflammatory and fibrotic diseases, offering an understanding of the individual components as potential treatments.
Collapse
Affiliation(s)
- Diana V Ávila-Martínez
- Laboratorio de Medicina Traslacional, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
- Doctorado en Ciencias de la Salud, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | - Wendy K Mixtega-Ruiz
- Laboratorio de Medicina Traslacional, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
- Doctorado en Ciencias Biológicas, Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | | | - Oscar Lopez-Franco
- Laboratorio de Medicina Traslacional, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
- Doctorado en Ciencias de la Salud, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | - Mónica Flores-Muñoz
- Laboratorio de Medicina Traslacional, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
- Doctorado en Ciencias de la Salud, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| |
Collapse
|
34
|
Keppeler K, Pesi A, Lange S, Helmstädter J, Strohm L, Ubbens H, Kuntić M, Kuntić I, Mihaliková D, Vujačić-Mirski K, Rosenberger A, Küster L, Frank C, Oelze M, Finger S, Zakrzewska A, Verdu E, Wild J, Karbach S, Wenzel P, Wild P, Leistner D, Münzel T, Daiber A, Schuppan D, Steven S. Vascular dysfunction and arterial hypertension in experimental celiac disease are mediated by gut-derived inflammation and oxidative stress. Redox Biol 2024; 70:103071. [PMID: 38354629 PMCID: PMC10876911 DOI: 10.1016/j.redox.2024.103071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024] Open
Abstract
AIMS We examined the cardiovascular effects of celiac disease (CeD) in a humanized mouse model, with a focus on vascular inflammation, endothelial dysfunction, and oxidative stress. METHODS AND RESULTS NOD.DQ8 mice genetically predisposed to CeD were subjected to a diet regime and oral gavage to induce the disease (gluten group vs. control). We tested vascular function, confirmed disease indicators, and evaluated inflammation and oxidative stress in various tissues. Plasma proteome profiling was also performed. CeD markers were confirmed in the gluten group, indicating increased blood pressure and impaired vascular relaxation. Pro-inflammatory genes were upregulated in this group, with increased CD11b+ myeloid cell infiltration and oxidative stress parameters observed in aortic and heart tissue. However, heart function remained unaffected. Plasma proteomics suggested the cytokine interleukin-17A (IL-17A) as a link between gut and vascular inflammation. Cardiovascular complications were reversed by adopting a gluten-free diet. CONCLUSION Our study sheds light in the heightened cardiovascular risk associated with active CeD, revealing a gut-to-cardiovascular inflammatory axis potentially mediated by immune cell infiltration and IL-17A. These findings augment our understanding of the link between CeD and cardiovascular disease providing clinically relevant insight into the underlying mechanism. Furthermore, our discovery that cardiovascular complications can be reversed by a gluten-free diet underscores a critical role for dietary interventions in mitigating cardiovascular risks associated with CeD.
Collapse
Affiliation(s)
- Karin Keppeler
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Aline Pesi
- Institute of Translational Immunology (TIM), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Simon Lange
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Johanna Helmstädter
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Lea Strohm
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Henning Ubbens
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Marin Kuntić
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Ivana Kuntić
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Dominika Mihaliková
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Ksenija Vujačić-Mirski
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Alexandra Rosenberger
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Leonie Küster
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Charlotte Frank
- Institute of Translational Immunology (TIM), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Stefanie Finger
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Agnieszka Zakrzewska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Elena Verdu
- Farncombe Digestive Disease Center, McMaster University, Hamilton, Canada
| | - Johannes Wild
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Susanne Karbach
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Philip Wenzel
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Philipp Wild
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - David Leistner
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany; Division of Cardiology, Goethe University Frankfurt, University Hospital, Department of Medicine III, Frankfurt a. M., Germany
| | - Thomas Münzel
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Andreas Daiber
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Detlef Schuppan
- Institute of Translational Immunology (TIM), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sebastian Steven
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Division of Cardiology, Goethe University Frankfurt, University Hospital, Department of Medicine III, Frankfurt a. M., Germany.
| |
Collapse
|
35
|
Kij A, Bar A, Czyzynska-Cichon I, Przyborowski K, Proniewski B, Mateuszuk L, Kurylowicz Z, Jasztal A, Buczek E, Kurpinska A, Suraj-Prazmowska J, Marczyk B, Matyjaszczyk-Gwarda K, Daiber A, Oelze M, Walczak M, Chlopicki S. Vascular protein disulfide isomerase A1 mediates endothelial dysfunction induced by angiotensin II in mice. Acta Physiol (Oxf) 2024; 240:e14116. [PMID: 38400621 DOI: 10.1111/apha.14116] [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: 12/17/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024]
Abstract
AIM Protein disulfide isomerases (PDIs) are involved in platelet aggregation and intravascular thrombosis, but their role in regulating endothelial function is unclear. Here, we characterized the involvement of vascular PDIA1 in angiotensin II (Ang II)-induced endothelial dysfunction in mice. METHODS Endothelial dysfunction was induced in C57BL/6JCmd male mice via Ang II subcutaneous infusion, and PDIA1 was inhibited with bepristat. Endothelial function was assessed in vivo with magnetic resonance imaging and ex vivo with a myography, while arterial stiffness was measured as pulse wave velocity. Nitric oxide (NO) bioavailability was measured in the aorta (spin-trapping electron paramagnetic resonance) and plasma (NO2 - and NO3 - levels). Oxidative stress, eNOS uncoupling (DHE-based aorta staining), and thrombin activity (thrombin-antithrombin complex; calibrated automated thrombography) were evaluated. RESULTS The inhibition of PDIA1 by bepristat in Ang II-treated mice prevented the impairment of NO-dependent vasodilation in the aorta as evidenced by the response to acetylcholine in vivo, increased systemic NO bioavailability and the aortic NO production, and decreased vascular stiffness. Bepristat's effect on NO-dependent function was recapitulated ex vivo in Ang II-induced endothelial dysfunction in isolated aorta. Furthermore, bepristat diminished the Ang II-induced eNOS uncoupling and overproduction of ROS without affecting thrombin activity. CONCLUSION In Ang II-treated mice, the inhibition of PDIA1 normalized the NO-ROS balance, prevented endothelial eNOS uncoupling, and, thereby, improved vascular function. These results indicate the importance of vascular PDIA1 in regulating endothelial function, but further studies are needed to elucidate the details of the mechanisms involved.
Collapse
Affiliation(s)
- Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Izabela Czyzynska-Cichon
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Kamil Przyborowski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Bartosz Proniewski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Lukasz Mateuszuk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Zuzanna Kurylowicz
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Elzbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Kurpinska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Joanna Suraj-Prazmowska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Brygida Marczyk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | | | - Andreas Daiber
- Laboratory of Molecular Cardiology, Department of Cardiology 1, The Center for Cardiology, University Medical Center, Mainz, Germany
| | - Matthias Oelze
- Laboratory of Molecular Cardiology, Department of Cardiology 1, The Center for Cardiology, University Medical Center, Mainz, Germany
| | - Maria Walczak
- Department of Toxicology, Jagiellonian University Medical College, Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Department of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
36
|
Ertuglu LA, Mutchler AP, Jamison S, Laffer CL, Saleem M, Blackwell DJ, Kryshtal DO, Sahinoz M, Sheng Q, Wanjalla CN, Pakala S, Justin Y, Gutierrez OM, Kleyman TR, Knollmann BC, Ikizler TA, Kirabo A. Eicosanoid-Regulated Myeloid ENaC and Isolevuglandin Formation in Human Salt-Sensitive Hypertension. Hypertension 2024; 81:516-529. [PMID: 37675576 PMCID: PMC10918035 DOI: 10.1161/hypertensionaha.123.21285] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/15/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND The mechanisms by which salt increases blood pressure in people with salt sensitivity remain unclear. Our previous studies found that high sodium enters antigen-presenting cells (APCs) via the epithelial sodium channel and leads to the production of isolevuglandins and hypertension. In the current mechanistic clinical study, we hypothesized that epithelial sodium channel-dependent isolevuglandin-adduct formation in APCs is regulated by epoxyeicosatrienoic acids (EETs) and leads to salt-sensitive hypertension in humans. METHODS Salt sensitivity was assessed in 19 hypertensive subjects using an inpatient salt loading and depletion protocol. Isolevuglandin-adduct accumulation in APCs was analyzed using flow cytometry. Gene expression in APCs was analyzed using cellular indexing of transcriptomes and epitopes by sequencing analysis of blood mononuclear cells. Plasma and urine EETs were measured using liquid chromatography-mass spectrometry. RESULTS Baseline isolevuglandin+ APCs correlated with higher salt-sensitivity index. Isolevuglandin+ APCs significantly decreased from salt loading to depletion with an increasing salt-sensitivity index. We observed that human APCs express the epithelial sodium channel δ subunit, SGK1 (salt-sensing kinase serum/glucocorticoid kinase 1), and cytochrome P450 2S1. We found a direct correlation between baseline urinary 14,15 EET and salt-sensitivity index, whereas changes in urinary 14,15 EET negatively correlated with isolevuglandin+ monocytes from salt loading to depletion. Coincubation with 14,15 EET inhibited high-salt-induced increase in isolevuglandin+ APC. CONCLUSIONS Isolevuglandin formation in APCs responds to acute changes in salt intake in salt-sensitive but not salt-resistant people with hypertension, and this may be regulated by renal 14,15 EET. Baseline levels of isolevuglandin+ APCs or urinary 14,15 EET may provide diagnostic tools for salt sensitivity without a protocol of salt loading.
Collapse
Affiliation(s)
- Lale A. Ertuglu
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ashley Pitzer Mutchler
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - S Jamison
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
- Meharry Medical College Nashville, Nashville, TN, United States
| | - Cheryl L. Laffer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Daniel J. Blackwell
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Dmytro O. Kryshtal
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Melis Sahinoz
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Department of Internal Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center Nashville, TN, USA
| | - Suman Pakala
- Department of Internal Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center Nashville, TN, USA
| | - Yu Justin
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Orlando M Gutierrez
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Thomas R. Kleyman
- Departments of Medicine, Cell Biology, Pharmacology, and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Björn C. Knollmann
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - T. Alp Ikizler
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
- Vanderbilt Center for Immunobiology (VCI)
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4)
- Vanderbilt Institute for Global Health (VIGH)
| |
Collapse
|
37
|
Benson LN, Mu S. Interferon gamma in the pathogenesis of hypertension - recent insights. Curr Opin Nephrol Hypertens 2024; 33:154-160. [PMID: 38164939 PMCID: PMC10842676 DOI: 10.1097/mnh.0000000000000966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
PURPOSE OF REVIEW The mounting body of evidence underscores the pivotal role of interferon gamma (IFNγ) in the pathogenesis of hypertension, prompting exploration of the mechanisms by which this cytokine fosters a pro-inflammatory immune milieu, subsequently exacerbating hypertension. In this review, we delve into recent preclinical and clinical studies from the past two years to elucidate how IFNγ participates in the progression of hypertension. RECENT FINDINGS IFNγ promotes renal CD8 + T cell accumulation by upregulating tubular PDL1 and MHC-I, intensifying cell-to-cell interaction. Intriguingly, a nucleotide polymorphism in LNK, predisposing towards hypertension, correlates with augmented T cell IFNγ production. Additionally, anti-IFNγ treatment exhibits protective effects against T cell-mediated inflammation during angiotensin II infusion or transverse aortic constriction. Moreover, knockout of the mineralocorticoid receptor in T cells protects against cardiac dysfunction induced by myocardial infarction, correlating with reduced IFNγ and IL-6, decreased macrophage recruitment, and attenuated fibrosis. Interestingly, increased IFNγ production correlates with elevated blood pressure, impacting individuals with type 2 diabetes, nondiabetics, and obese hypertensive patients. SUMMARY These revelations spotlight IFNγ as the critical mediator bridging the initial phase of blood pressure elevation with the sustained and exacerbated pathology. Consequently, blocking IFNγ signaling emerges as a promising therapeutic target to improve the management of this 'silent killer.'
Collapse
Affiliation(s)
- Lance N. Benson
- Heersink School of Medicine: Department of CardioRenal Physiology and Medicine, Division of Nephrology University of Alabama at Birmingham, Birmingham, Alabama
| | - Shengyu Mu
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
38
|
Gan L, Ye D, Feng Y, Pan H, Lu X, Wan J, Ye J. Immune cells and hypertension. Immunol Res 2024; 72:1-13. [PMID: 38044398 DOI: 10.1007/s12026-023-09414-z] [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: 12/07/2022] [Accepted: 08/10/2023] [Indexed: 12/05/2023]
Abstract
Hypertension is one of the leading causes of death due to target organ injury from cardiovascular disease. Although there are many treatments, only one-sixth of hypertensive patients effectively control their blood pressure. Therefore, further understanding the pathogenesis of hypertension is essential for the treatment of hypertension. Much research shows that immune cells play an important role in the pathogenesis of hypertension. Here, we discuss the roles of different immune cells in hypertension. Many immune cells participate in innate and adaptive immune responses, such as monocytes/macrophages, neutrophils, dendritic cells, NK cells, and B and T lymphocytes. Immune cells infiltrate the blood vessels, kidneys, and hearts and cause damage. The mechanism is that immune cells secrete cytokines such as interleukin, interferon, and tumor necrosis factor, which affect the inflammatory reaction, oxidative stress, and kidney sodium water retention, and finally aggravate or reduce the dysfunction, remodeling, and fibrosis of the blood vessel, kidney, and heart to participate in blood pressure regulation. This article reviews the research progress on immune cells and hypertension.
Collapse
Affiliation(s)
- Liren Gan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiyi Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
39
|
Gonzalez AL, Dungan MM, Smart CD, Madhur MS, Doran AC. Inflammation Resolution in the Cardiovascular System: Arterial Hypertension, Atherosclerosis, and Ischemic Heart Disease. Antioxid Redox Signal 2024; 40:292-316. [PMID: 37125445 PMCID: PMC11071112 DOI: 10.1089/ars.2023.0284] [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: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023]
Abstract
Significance: Chronic inflammation has emerged as a major underlying cause of many prevalent conditions in the Western world, including cardiovascular diseases. Although targeting inflammation has emerged as a promising avenue by which to treat cardiovascular disease, it is also associated with increased risk of infection. Recent Advances: Though previously assumed to be passive, resolution has now been identified as an active process, mediated by unique immunoresolving mediators and mechanisms designed to terminate acute inflammation and promote tissue repair. Recent work has determined that failures of resolution contribute to chronic inflammation and the progression of human disease. Specifically, failure to produce pro-resolving mediators and the impaired clearance of dead cells from inflamed tissue have been identified as major mechanisms by which resolution fails in disease. Critical Issues: Drawing from a rapidly expanding body of experimental and clinical studies, we review here what is known about the role of inflammation resolution in arterial hypertension, atherosclerosis, myocardial infarction, and ischemic heart disease. For each, we discuss the involvement of specialized pro-resolving mediators and pro-reparative cell types, including T regulatory cells, myeloid-derived suppressor cells, and macrophages. Future Directions: Pro-resolving therapies offer the promise of limiting chronic inflammation without impairing host defense. Therefore, it is imperative to better understand the mechanisms underlying resolution to identify therapeutic targets. Antioxid. Redox Signal. 40, 292-316.
Collapse
Affiliation(s)
- Azuah L. Gonzalez
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Matthew M. Dungan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - C. Duncan Smart
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Meena S. Madhur
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Amanda C. Doran
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
40
|
Sørensen M, Pershagen G, Thacher JD, Lanki T, Wicki B, Röösli M, Vienneau D, Cantuaria ML, Schmidt JH, Aasvang GM, Al-Kindi S, Osborne MT, Wenzel P, Sastre J, Fleming I, Schulz R, Hahad O, Kuntic M, Zielonka J, Sies H, Grune T, Frenis K, Münzel T, Daiber A. Health position paper and redox perspectives - Disease burden by transportation noise. Redox Biol 2024; 69:102995. [PMID: 38142584 PMCID: PMC10788624 DOI: 10.1016/j.redox.2023.102995] [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: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 12/26/2023] Open
Abstract
Transportation noise is a ubiquitous urban exposure. In 2018, the World Health Organization concluded that chronic exposure to road traffic noise is a risk factor for ischemic heart disease. In contrast, they concluded that the quality of evidence for a link to other diseases was very low to moderate. Since then, several studies on the impact of noise on various diseases have been published. Also, studies investigating the mechanistic pathways underlying noise-induced health effects are emerging. We review the current evidence regarding effects of noise on health and the related disease-mechanisms. Several high-quality cohort studies consistently found road traffic noise to be associated with a higher risk of ischemic heart disease, heart failure, diabetes, and all-cause mortality. Furthermore, recent studies have indicated that road traffic and railway noise may increase the risk of diseases not commonly investigated in an environmental noise context, including breast cancer, dementia, and tinnitus. The harmful effects of noise are related to activation of a physiological stress response and nighttime sleep disturbance. Oxidative stress and inflammation downstream of stress hormone signaling and dysregulated circadian rhythms are identified as major disease-relevant pathomechanistic drivers. We discuss the role of reactive oxygen species and present results from antioxidant interventions. Lastly, we provide an overview of oxidative stress markers and adverse redox processes reported for noise-exposed animals and humans. This position paper summarizes all available epidemiological, clinical, and preclinical evidence of transportation noise as an important environmental risk factor for public health and discusses its implications on the population level.
Collapse
Affiliation(s)
- Mette Sørensen
- Work, Environment and Cancer, Danish Cancer Institute, Copenhagen, Denmark; Department of Natural Science and Environment, Roskilde University, Denmark.
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jesse Daniel Thacher
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Timo Lanki
- Department of Health Security, Finnish Institute for Health and Welfare, Kuopio, Finland; School of Medicine, University of Eastern Finland, Kuopio, Finland; Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Benedikt Wicki
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Martin Röösli
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Danielle Vienneau
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Manuella Lech Cantuaria
- Work, Environment and Cancer, Danish Cancer Institute, Copenhagen, Denmark; Research Unit for ORL - Head & Neck Surgery and Audiology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Jesper Hvass Schmidt
- Research Unit for ORL - Head & Neck Surgery and Audiology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Gunn Marit Aasvang
- Department of Air Quality and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | - Sadeer Al-Kindi
- Department of Medicine, University Hospitals, Harrington Heart & Vascular Institute, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Michael T Osborne
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA, USA; Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Philip Wenzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Spain
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt Am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - Rainer Schulz
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, 35392, Gießen, Germany
| | - Omar Hahad
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Marin Kuntic
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Faculty of Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Katie Frenis
- Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
41
|
Dinh QN, Lo C, Zhang DW, Tran V, Gibson-Hughes T, Sheriff A, Diep H, Kim HA, Zhang SR, Barreto-Arce LJ, Jelinic M, Vinh A, Arumugam TV, Chan ST, Lim R, Drummond GR, Sobey CG, De Silva TM. Human amnion epithelial cell therapy reduces hypertension-induced vascular stiffening and cognitive impairment. Sci Rep 2024; 14:1837. [PMID: 38246932 PMCID: PMC10800338 DOI: 10.1038/s41598-024-52214-0] [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: 11/17/2023] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
Vascular inflammation and fibrosis are hallmarks of hypertension and contribute to the development of cardiovascular disease and cognitive impairment. However, current anti-hypertensive drugs do not treat the underlying tissue damage, such as inflammation-associated fibrosis. Human amnion epithelial cells have several properties amenable for treating vascular pathology. This study tested the effect of amnion epithelial cells on vascular pathology and cognitive impairment during hypertension. Male C57Bl6 mice (8-12 weeks) were administered vehicle (saline; n = 58) or angiotensin II (0.7 mg/kg/d, n = 56) subcutaneously for 14 d. After surgery, a subset of mice were injected with 106 amnion epithelial cells intravenously. Angiotensin II infusion increased systolic blood pressure, aortic pulse wave velocity, accumulation of aortic leukocytes, and aortic mRNA expression of collagen subtypes compared to vehicle-infused mice (n = 9-11, P < 0.05). Administration of amnion epithelial cells attenuated these effects of angiotensin II (P < 0.05). Angiotensin II-induced cognitive impairment was prevented by amnion epithelial cell therapy (n = 7-9, P < 0.05). In the brain, amnion epithelial cells modulated some of the inflammatory genes that angiotensin II promoted differential expression of (n = 6, p-adjusted < 0.05). These findings suggest that amnion epithelial cells could be explored as a potential therapy to inhibit vascular pathology and cognitive impairment during hypertension.
Collapse
Affiliation(s)
- Quynh Nhu Dinh
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Cecilia Lo
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - David Wong Zhang
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Vivian Tran
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Tayla Gibson-Hughes
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Ashleigh Sheriff
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Henry Diep
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Hyun Ah Kim
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Shenpeng R Zhang
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Liz J Barreto-Arce
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Maria Jelinic
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Antony Vinh
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Thiruma V Arumugam
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Siow Teng Chan
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Rebecca Lim
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Grant R Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Christopher G Sobey
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia.
| | - T Michael De Silva
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia.
| |
Collapse
|
42
|
Perrotta S, Carnevale D. Brain-Splenic Immune System Interactions in Hypertension: Cellular and Molecular Mechanisms. Arterioscler Thromb Vasc Biol 2024; 44:65-75. [PMID: 37942610 DOI: 10.1161/atvbaha.123.318230] [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: 04/07/2023] [Accepted: 10/20/2023] [Indexed: 11/10/2023]
Abstract
Hypertension represents a major worldwide cause of death and disability, and it is becoming increasingly clear that available therapies are not sufficient to reduce the risk of major cardiovascular events. Various mechanisms contribute to blood pressure increase: neurohormonal activation, autonomic nervous system imbalance, and immune activation. Of note, the brain is an important regulator of blood pressure levels; it recognizes the peripheral perturbation and organizes a reflex response by modulating immune system and hormonal release to attempt at restoring the homeostasis. The connection between the brain and peripheral organs is mediated by the autonomic nervous system, which also modulates immune and inflammatory responses. Interestingly, an increased autonomic nervous system activity has been correlated with an altered immune response in cardiovascular diseases. The spleen is the largest immune organ exerting a potent influence on the cardiovascular system during disease and is characterized by a dense noradrenergic innervation. Taken together, these aspects led to hypothesize a key role of neuroimmune mechanisms in the onset and progression of hypertension. This review discusses how the nervous and splenic immune systems interact and how the mechanisms underlying the neuroimmune cross talk influence the disease progression.
Collapse
Affiliation(s)
- Sara Perrotta
- Department of Angiocardioneurology and Translational Medicine, Unit of Neuro and Cardiovascular Pathophysiology, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, Pozzilli, Italy (S.P., D.C.)
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, Unit of Neuro and Cardiovascular Pathophysiology, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, Pozzilli, Italy (S.P., D.C.)
- Department of Molecular Medicine, "Sapienza" University of Rome, Italy (D.C.)
| |
Collapse
|
43
|
Zhang J, Tu Y, Wei J, Zheng R, Shao J, Chen Q, Liang G, Ying H, Han X, Shi Q. Dectin1 contributes to hypertensive vascular injury by promoting macrophage infiltration through activating the Syk/NF-κB pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166911. [PMID: 37813169 DOI: 10.1016/j.bbadis.2023.166911] [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: 06/13/2023] [Revised: 09/13/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023]
Abstract
Vascular injury is an early manifestation leading to end-organ damage in hypertension pathogenesis, which involves a macrophage-associated immune response. Dendritic cell-associated C-type lectin-1 (Dectin1) is a pivotal player in regulating inflammation-mediated cardiovascular disease. However, its role in hypertension-induced vascular damage and the underlying mechanisms remain unclear. We hypothesized that Dectin1 might accelerate angiotensin II (Ang II)- or deoxycorticosterone acetate-salt (DOCA-salt)-induced vascular injury through proinflammatory actions in macrophages. Macrophage Dectin1 was upregulated in mouse aortic tissues stimulated with Ang II. In the peripheral blood, Ang II also increased CD11b+F4/80+ macrophages in mice. In our constructed Dectin1 knockout mice, Dectin1 deletion protected against Ang II-induced EB extravasation and aortic wall thickness. Deficiency of Dectin1 or its pharmacological inhibition considerably improved fibrosis and inflammation responses, accompanied by a reduction in M1 macrophage polarization as well as proinflammatory cytokines and chemokines induced by Ang II or DOCA-salt. Through the bone marrow (BM) transplantation assay, these effects were verified in the wild type mice reconstituted with Dectin1-deficient BM cells. Mechanistically, Ang II promoted Dectin1 homodimerization, thereby triggering the spleen tyrosine kinase/nuclear factor kappa B pro-inflammatory cascade to induce the expression of inflammatory factors and chemokines in vivo and in vitro. In conclusion, Dectin1 has an essential role in the pathogenic procedure of Ang II-stimulated or DOCA-salt-induced vascular damage in mice and represents a promising therapeutic target for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiajia Zhang
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China; Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou 310013, China
| | - Yu Tu
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China; Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou 310013, China
| | - Jiajia Wei
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China
| | - Ruyi Zheng
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China
| | - Ji Shao
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China
| | - Qinhua Chen
- Key Laboratory of TCM Clinical Pharmacy, Shenzhen Baoan Authentic TCM Therapy Hospital, Guangzhou University of Chinese Medicine, Shenzhen 518101, China
| | - Guang Liang
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China
| | - Huazhong Ying
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China; Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou 310013, China.
| | - Xue Han
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China.
| | - Qiaojuan Shi
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China; Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou 310013, China.
| |
Collapse
|
44
|
Zhang YL, Bai J, Yu WJ, Lin QY, Li HH. CD11b mediates hypertensive cardiac remodeling by regulating macrophage infiltration and polarization. J Adv Res 2024; 55:17-31. [PMID: 36822392 PMCID: PMC10770112 DOI: 10.1016/j.jare.2023.02.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
INTRODUCTION Leukocyte infiltration is an early event during cardiac remodeling frequently leading to heart failure (HF). Integrins mediate leukocyte infiltration during inflammation. However, the importance of specific integrins in hypertensive cardiac remodeling is still unclear. OBJECTIVES To elucidate the significance of CD11b in hypertensive cardiac remodeling. METHODS Angiotensin (Ang II) or deoxycorticosterone acetate (DOCA)-salt was used to induce cardiac remodeling in mice of gene knockout (KO), bone marrow (BM) chimera, and the CD11b neutralizing antibody or agonist leukadherin-1 (LA1) treatment. RESULTS Our microarray data showed that integrin subunits Itgam (CD11b) and Itgb2 (CD18) were the most highly upregulated in Ang II-infused hearts. CD11b expression and CD11b/CD18+ myelomonocytes were also time-dependently increased. KO or pharmacological blockade of CD11b greatly attenuated cardiac remodeling and macrophage infiltration and M1 polarization induced by Ang II or DOCA-salt. This protection was verified in wild-type mice transplanted with CD11b-deficient BM cells. Conversely, administration of CD11b agonist LA1 showed the opposite effects. Further, CD11b KO reduced Ang II-induced macrophage adhesion and M1 polarization, leading to reduction of cardiomyocyte enlargement and fibroblast differentiation in vitro. The numbers of CD14+CD11b+CD18+ monocytes and CD15+CD11b+CD18+ granulocytes were obviously higher in HF patients than in normal controls. CONCLUSION Our data demonstrate an important role of CD11b+ myeloid cells in hypertensive cardiac remodeling, and suggest that HF may benefit from targeting CD11b.
Collapse
Affiliation(s)
- Yun-Long Zhang
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Jie Bai
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, Dalian 116011, China
| | - Wei-Jia Yu
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, Dalian 116011, China
| | - Qiu-Yue Lin
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, Dalian 116011, China.
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China.
| |
Collapse
|
45
|
Zhang J, Yin Z, Xu Y, Wei C, Peng S, Zhao M, Liu J, Xu S, Pan W, Zheng Z, Liu S, Ye J, Qin JJ, Wan J, Wang M. Resolvin E1/ChemR23 Protects Against Hypertension and Vascular Remodeling in Angiotensin II-Induced Hypertensive Mice. Hypertension 2023; 80:2650-2664. [PMID: 37800344 DOI: 10.1161/hypertensionaha.123.21348] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 09/18/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Inflammation plays a critical role in the development of hypertension and vascular remodeling. Resolvin E1 (RvE1), as one of the specialized proresolving lipid mediators, promotes inflammation resolution by binding with a G protein-coupled receptor, ChemR23 (chemerin receptor 23). However, whether RvE1/ChemR23 regulates hypertension and vascular remodeling is unknown. METHODS Hypertension in mice was induced by Ang II (angiotensin II) infusion (750 ng/kg per minute), and RvE1 (2 µg/kg per day) was administered through intraperitoneal injection. Loss of ChemR23 was achieved by mice receiving intravenous injection of adeno-associated virus 9-encoding shRNA against ChemR23. RESULTS Aortic ChemR23 expression was increased in Ang II-induced hypertensive mice and that ChemR23 was mainly expressed on vascular smooth muscle cells (VSMCs). RvE1 lowered blood pressure, reduced aortic media thickness, attenuated aortic fibrosis, and mitigated VSMC phenotypic transformation and proliferation in hypertensive mice, which were all reversed by the knockdown of ChemR23. Moreover, RvE1 reduced the aortic infiltration of macrophages and T cells, which was also reversed by ChemR23 knockdown. RvE1 inhibited Ccl5 expression in VSMCs via the AMPKα (AMP-activated protein kinase α)/Nrf2 (nuclear factor E2-related factor 2)/canonical NF-κB (nuclear factor κB) pathway, thereby reducing the infiltration of macrophages and T cells. The AMPKα/Nrf2 pathway also mediated the effects of RvE1 on VSMC phenotypic transformation and proliferation. In patients with hypertension, the serum levels of RvE1 and other eicosapentaenoic acid-derived metabolites were significantly decreased. CONCLUSIONS RvE1/ChemR23 ameliorated hypertension and vascular remodeling by activating AMPKα/Nrf2 signaling, which mediated immune cell infiltration by inhibiting the canonical NF-κB/Ccl5 pathway, and regulated VSMC proliferation and phenotypic transformation. RvE1/ChemR23 may be a potential therapeutic target for hypertension.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Yao Xu
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Wei Pan
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Siqi Liu
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Jing Ye
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Center for Healthy Aging, Wuhan University School of Nursing, China (J.-J.Q.)
| | - Jun Wan
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| |
Collapse
|
46
|
Barhoumi T, Todryk S. Role of monocytes/macrophages in renin-angiotensin system-induced hypertension and end organ damage. Front Physiol 2023; 14:1199934. [PMID: 37854465 PMCID: PMC10579565 DOI: 10.3389/fphys.2023.1199934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/12/2023] [Indexed: 10/20/2023] Open
Abstract
The renin-angiotensin system (RAS) is a central modulator of cardiovascular physiology. Pathophysiology of hypertension is commonly accompanied by hyper-activation of RAS. Angiotensin II receptor blockers (ARBs) and Angiotensin-converting enzyme (ACE) inhibitors are the gold standard treatment for hypertension. Recently, several studies highlighted the crucial role of immune system in hypertension. Angiotensin-II-induced hypertension is associated with low grade inflammation characterized by innate and adaptive immune system dysfunction. Throughout the progression of hypertension, monocyte/macrophage cells appear to have a crucial role in vascular inflammation and interaction with the arterial wall. Since myelomonocytic cells potentially play a key role in angiotensin-II-induced hypertension and organ damage, pharmacological targeting of RAS components in monocyte/macrophages may possibly present an innovative strategy for treatment of hypertension and related pathology.
Collapse
Affiliation(s)
- Tlili Barhoumi
- Medical Research Core Facility and Platforms (MRCFP), King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Stephen Todryk
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
47
|
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: 2] [Impact Index Per Article: 1.0] [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
|
48
|
Ribeiro Vitorino T, Ferraz do Prado A, Bruno de Assis Cau S, Rizzi E. MMP-2 and its implications on cardiac function and structure: Interplay with inflammation in hypertension. Biochem Pharmacol 2023; 215:115684. [PMID: 37459959 DOI: 10.1016/j.bcp.2023.115684] [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: 05/27/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023]
Abstract
Hypertension is one of the leading risk factors for the development of heart failure. Despite being a multifactorial disease, in recent years, preclinical and clinical studies suggest strong evidence of the pivotal role of inflammatory cells and cytokines in the remodeling process and cardiac dysfunction. During the heart remodeling, activation of extracellular matrix metalloproteinases (MMPs) occurs, with MMP-2 being one of the main proteases secreted by cardiomyocytes, fibroblasts, endothelial and inflammatory cells in cardiac tissue. In this review, we will address the process of cardiac remodeling and injury induced by the increase in MMP-2 and the main signaling pathways involving cytokines and inflammatory cells in the process of transcriptional, secretion and activation of MMP-2. In addition, an interaction and coordinated action between MMP-2 and inflammation are explored and significant in maintaining the cardiac cycle. These observations suggest that new therapeutic opportunities targeting MMP-2 could be used to reduce inflammatory biomarkers and reduce cardiac damage in hypertension.
Collapse
Affiliation(s)
- Thaís Ribeiro Vitorino
- Unit of Biotechnology, University of Ribeirao Preto, UNAERP, Brazil; Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, UNICAMP, Brazil
| | - Alejandro Ferraz do Prado
- Cardiovascular System Pharmacology and Toxicology, Institute of Biological Sciences, Federal University of Para, UFPA, Brazil
| | - Stefany Bruno de Assis Cau
- Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, UFMG, Brazil.
| | - Elen Rizzi
- Unit of Biotechnology, University of Ribeirao Preto, UNAERP, Brazil.
| |
Collapse
|
49
|
Daou D, Gillette TG, Hill JA. Inflammatory Mechanisms in Heart Failure with Preserved Ejection Fraction. Physiology (Bethesda) 2023; 38:0. [PMID: 37013947 PMCID: PMC10396273 DOI: 10.1152/physiol.00004.2023] [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: 01/18/2023] [Revised: 03/27/2023] [Accepted: 04/02/2023] [Indexed: 04/05/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is now the most common form of heart failure and a significant public health concern for which limited effective therapies exist. Inflammation triggered by comorbidity burden is a critical element of HFpEF pathophysiology. Here, we discuss evidence for comorbidity-driven systemic and myocardial inflammation and the mechanistic role of inflammation in pathological myocardial remodeling in HFpEF.
Collapse
Affiliation(s)
- Daniel Daou
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Thomas G Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
50
|
Yang Y, Karampoor S, Mirzaei R, Borozdkin L, Zhu P. The interplay between microbial metabolites and macrophages in cardiovascular diseases: A comprehensive review. Int Immunopharmacol 2023; 121:110546. [PMID: 37364331 DOI: 10.1016/j.intimp.2023.110546] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/11/2023] [Accepted: 06/18/2023] [Indexed: 06/28/2023]
Abstract
The gut microbiome has emerged as a crucial player in developing and progressing cardiovascular diseases (CVDs). Recent studies have highlighted the role of microbial metabolites in modulating immune cell function and their impact on CVD. Macrophages, which have a significant function in the pathogenesis of CVD, are very vulnerable to the effects of microbial metabolites. Microbial metabolites, such as short-chain fatty acids (SCFAs) and trimethylamine-N-oxide (TMAO), have been linked to atherosclerosis and the regulation of immune functions. Butyrate has been demonstrated to reduce monocyte migration and inhibit monocyte attachment to injured endothelial cells, potentially contributing to the attenuation of the inflammatory response and the progression of atherosclerosis. On the other hand, TMAO, another compound generated by gut bacteria, has been linked to atherosclerosis due to its impact on lipid metabolism and the accumulation of cholesterol in macrophages. Indole-3-propionic acid, a tryptophan metabolite produced solely by microbes, has been found to promote the development of atherosclerosis by stimulating macrophage reverse cholesterol transport (RCT) and raising the expression of ABCA1. This review comprehensively discusses how various microbiota-produced metabolites affect macrophage polarization, inflammation, and foam cell formation in CVD. We also highlight the mechanisms underlying these effects and the potential therapeutic applications of targeting microbial metabolites in treating CVD.
Collapse
Affiliation(s)
- Yongzheng Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Leonid Borozdkin
- Department of Maxillofacial Surgery, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510100, China.
| |
Collapse
|