1
|
Battisha A, Kahlon A, Kalra DK. Sleep-Disordered Breathing and Hypertension-A Systematic Review. J Clin Med 2025; 14:3115. [PMID: 40364148 PMCID: PMC12072724 DOI: 10.3390/jcm14093115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Sleep-disordered breathing (SDB), historically referred to as "sleep apnea syndrome", particularly obstructive sleep apnea (OSA), is an independent risk factor for hypertension (HTN), stroke, heart failure, arrhythmias, and other cardiovascular disorders. Despite the well-established link between OSA and HTN and its high occurrence in cardiovascular disorders, the focus on the complex OSA-HTN axis is often overlooked or inadequately managed, which might explain the lack of notable improvements in cardiovascular outcomes for this patient population. Understanding the complex relationship between OSA and HTN is crucial due to its significant implications for clinical practice and public health. Methods: Using an expanded list of relevant MeSH terms, including "sleep-disordered breathing" and "sleep apnea syndrome", and following the PRISMA model, peer-reviewed articles were systematically selected. Studies published from January 2000 through December 2024 were identified and screened based on predefined inclusion and exclusion criteria. Results: This review emphasizes both OSA's independent and interaction effects on cardiovascular health and outcomes across different populations. It identifies key factors mediating the association between OSA and HTN. Conclusions: Multimodal management, including continuous positive airway pressure and lifestyle modification, is essential for treating hypertension related to OSA. Effective management of the OSA-HTN relationship is vital to improving cardiovascular outcomes.
Collapse
Affiliation(s)
| | | | - Dinesh K. Kalra
- Division of Cardiology, Department of Medicine, University of Louisville, 201 Abraham Flexner Way, Suite 600, Louisville, KY 40202, USA; (A.B.); (A.K.)
| |
Collapse
|
2
|
Shao Q, Wang H, Li S, Zeng M, Zhang S, Yan X. IRF5 Mediates Artery Inflammation in Salt-Sensitive Hypertension by Regulating STAT1 and STAT2 Phosphorylation to Increase ESM1 Transcription: Insights from Bioinformatics and Mechanistic Analysis. Int J Mol Sci 2025; 26:3722. [PMID: 40332339 PMCID: PMC12027925 DOI: 10.3390/ijms26083722] [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: 02/15/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
Salt-sensitive hypertension (SSH) is closely associated with arterial inflammation, yet its molecular mechanisms remain unclear. In this study, we utilized deoxycorticosterone acetate (DOCA)-salt-induced hypertensive mice, which exhibited elevated blood pressure and significant arterial inflammation. Single-cell RNA sequencing (scRNA-seq) identified interferon regulatory factor 5 (IRF5) and its downstream targets, signal transducer and activator of transcription (STAT), as key regulators of these inflammatory changes. In vivo, IRF5 levels were significantly elevated in the DOCA group, while STAT1 and STAT2 protein levels were comparable to those in the normal salt group. However, nuclear levels of phosphorylated STAT1 (pSTAT1) and phosphorylated STAT2 (pSTAT2) were markedly higher in the DOCA group. Furthermore, scRNA-seq analysis showed increased IRF5 expression in endothelial cells (ECs) in both human and mouse aorta samples. In vitro, IRF5 knockdown in artery ECs led to a reduction in nuclear pSTAT1 and pSTAT2 expression. These results suggest that IRF5 promotes STAT1 and STAT2 phosphorylation, enabling their nuclear translocation. Additionally, RNA sequencing indicated a positive correlation between endothelial cell-specific molecule 1 (ESM1) and STAT1/STAT2. Using the UCSC and JASPAR databases, we identified multiple binding sites for the STAT1::STAT2 dimer on the ESM1 promoter. Luciferase reporter assays revealed enhanced ESM1 transcription following pSTAT1::pSTAT2 binding, and pinpoint potential binding sites. Chromatin Immunoprecipitation Quantitative PCR (ChIP-qPCR) further confirmed the specific binding sites between the pSTAT1::pSTAT2 dimer and the ESM1 promoter. These findings highlight the critical role of the IRF5-pSTAT1::pSTAT2-ESM1 pathway in the pathogenesis of SSH and suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Qiaoyu Shao
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hao Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
| | - Shicheng Li
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
| | - Mengying Zeng
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College Hospital, Beijing 100730, China
| | - Xiaowei Yan
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
| |
Collapse
|
3
|
Xu Z, Yu H, Zhuang R, Fan Q. Immunotherapy for hypertensive end-organ damage: a new therapeutic strategy. Essays Biochem 2025; 0:EBC20243000. [PMID: 40134277 DOI: 10.1042/ebc20243000] [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: 12/19/2024] [Accepted: 03/07/2025] [Indexed: 03/27/2025]
Abstract
Hypertension represents a highly prevalent chronic condition and stands among the foremost contributors to premature mortality on a global scale. Its etiopathogenesis is intricate and multifaceted, being shaped by a diverse array of elements such as age, genetic predisposition, and activation of the neuroendocrine apparatus. Mounting evidence has shed light on the significant part that autoimmune responses play in hypertension and the ensuing damage to end organs. Virtually all varieties of immune cells, spanning both innate and adaptive immune compartments, exhibit a close correlation with the progression of hypertension. These immune cells infiltrate the kidney and vascular mesenchyme, subsequently discharging potent cytokines, reactive oxygen species, and metalloproteinases. This cascade of events can affect the functionality of local blood vessels and potentially precipitate adverse structural and functional alterations in crucial organs like the heart and kidney. In recent times, the management of end-organ damage in hypertension has emerged as a pivotal scientific focus. A multitude of researchers are actively engaged in probing efficacious intervention regimens, among which immunotherapy strategies hold considerable promise and anticipation as a prospective avenue.
Collapse
Affiliation(s)
- Zhiyang Xu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210000, China
- Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Haisheng Yu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210000, China
| | - Rulin Zhuang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Qin Fan
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210000, China
| |
Collapse
|
4
|
Zhang X, Wang G, Li M, Li Y, Luo X, Liu Y, Zhang X, Hocher JG, Krämer BK, Hocher B, Yang X. Both partial inactivation as well as activation of NF-κB signaling lead to hypertension and chronic kidney disease. Nephrol Dial Transplant 2024; 39:1993-2004. [PMID: 38614958 DOI: 10.1093/ndt/gfae090] [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: 10/26/2023] [Indexed: 04/15/2024] Open
Abstract
BACKGROUND Activation of nuclear factor-kappa B (NF-κB) signalling is key in the pathogenesis of chronic kidney disease (CKD). However, a certain level of NF-κB activity is necessary to enable tissue repair. METHODS The relationship between activated and inactivated NF-κB signaling and the pathogenesis of CKD was investigated using mouse models of NF-κB partial inactivation (mutating cysteine at position 59 of the sixth exon on the NF-κB gene into alanine) and activation (mutating cysteine at position 59 of the sixth exon on the NF-κB gene into serine). RESULTS The density of CD3, CD8, CD68 positive cells, as well as the expression of interleukin 6, Tumor necrosis factor receptor associated factor 1 and Nef-associated factor 1 in the kidney tissues of NF-κBC59A mice were reduced, whereas an opposing pattern was observed in the NF-κBC59S mice. Blood pressure, kidney fibrosis (analyzed by periodic acid-Schiff, Masson trichrome and Sirius Red staining, as well as α-SMA immunofluorescence), serum creatinine and urinary albumin-to-creatinine ratio are markedly increased in NF-κB-activated and -inactivated mice compared with controls. Transmission electron microscopy indicated that the glomerular basement membrane was thicker in both NF-κBC59A and NF-κBC59S mice compared with wild-type mice. CONCLUSIONS Using mice models with partially activated and inactivated NF-κB pathways suggests that there is an apparently U-shaped relationship between blood pressure, kidney function as well as morphology and the activation of the NF-κB pathway. A certain optimal activity of the NF-κB pathway seems to be important to maintain optimal kidney function and morphology.
Collapse
Affiliation(s)
- Xiaotan Zhang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
- Department of Pathology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Ming Li
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Yunjin Li
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Xin Luo
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Yvonne Liu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology, Pneumology), University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
- Medical Faculty of Charité University Berlin, Berlin, Germany
| | - Xiaoli Zhang
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology, Pneumology), University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Johann-Georg Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology, Pneumology), University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
- Second Medical Faculty, Charles University Prague, Prague, Czech Republic
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology, Pneumology), University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology, Pneumology), University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
- Reproductive, Genetic Hospital of CITIC-Xiangya, Changsha, China, Institute of Medical Diagnostics, IMD, Berlin, Germany
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center, Clifford Hospital, Guangzhou, China
| |
Collapse
|
5
|
Visca D, Ardesi F, Zappa M, Pignatti P, Grossi S, Vanetti M, Migliori GB, Centis R, Angeli F, Spanevello A. Asthma and hypertension: the role of airway inflammation. Front Med (Lausanne) 2024; 11:1451625. [PMID: 39450103 PMCID: PMC11499200 DOI: 10.3389/fmed.2024.1451625] [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: 06/19/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Asthma is a chronic inflammatory respiratory disease often associated with comorbidities. Among cardiovascular comorbidities, arterial hypertension seems to create an additional health burden in asthmatics. However, evidence on this relationship is lacking. Objective Our study aims to evaluate the characteristics of hypertensive asthmatics, focusing on the role of inflammation as a possible link between these diseases. Methods We conducted a monocentric retrospective analysis consecutively including asthmatics who underwent induced sputum (IS) at our asthma referral center. Patients were divided in two groups according to presence or absence of history of hypertension. Clinical, functional, and inflammatory (airway and systemic) data were collected. Results Data on two hundred and sixty asthmatic patients were analyzed. Seventy-nine (30.4%) of them had a diagnosis of hypertension requiring a specific pharmacological treatment. Asthmatics with hypertension were more frequently male (p = 0.047), older (p < 0.001), and with higher body max index (BMI) (p < 0.001) when compared to normotensive patients. No difference concerning asthma control, severity and pharmacological treatment was observed between the two groups (all p > 0.05); distribution of comorbidities and lung function impairment (forced expiratory volume in the first second (FEV1) and forced vital capacity (FVC); all p < 0.05) were statistically different between groups. Mixed granulocytic airway inflammation was prevalent in the hypertensive asthmatics (p = 0.014). Interestingly, a multivariable analysis revealed that age ≥ 65 years and an increased percentage of sputum neutrophils (≥61%) were independent predictors of hypertensive status (p < 0.001). Conclusion Our data suggest that neutrophilic airway inflammation (as evaluated by induced sputum) is strictly associated with hypertension. In clinical practice, phenotyping asthmatic patients with comorbidities like hypertension could be useful also from a therapeutic point of view. Additional studies are mandatory to further elucidate the role of neutrophilic airway inflammation in asthma with cardiovascular diseases.
Collapse
Affiliation(s)
- Dina Visca
- Department of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Francesco Ardesi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Martina Zappa
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Patrizia Pignatti
- Allergy and Immunology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Sarah Grossi
- Department of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Marco Vanetti
- Department of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Giovanni Battista Migliori
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Rosella Centis
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Fabio Angeli
- Department of Medicine and Technological Innovation (DiMIT), University of Insubria, Varese, Italy
- Department of Medicine and Cardiopulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Antonio Spanevello
- Department of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
6
|
Li X, Ding H, Feng G, Huang Y. Role of angiotensin converting enzyme in pathogenesis associated with immunity in cardiovascular diseases. Life Sci 2024; 352:122903. [PMID: 38986897 DOI: 10.1016/j.lfs.2024.122903] [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: 01/19/2024] [Revised: 06/18/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024]
Abstract
Angiotensin converting enzyme (ACE) is not only a critical component in the renin-angiotensin system (RAS), but also suggested as an important mediator for immune response and activity, such as immune cell mobilization, metabolism, biogenesis of immunoregulatory molecules, etc. The chronic duration of cardiovascular diseases (CVD) has been increasingly considered to be triggered by uncontrolled pathologic immune reactions from myeloid cells and lymphocytes. Considering the potential anti-inflammatory effect of the traditional antihypertensive ACE inhibitor (ACEi), we attempt to elucidate whether ACE and its catalytically relevant substances as well as signaling pathways play a role in the immunity-related pathogenesis of common CVD, such as arterial hypertension, atherosclerosis and arrythmias. ACEi was also reported to benefit the prognoses of COVID-19-positive patients with CVD, and COVID-19 disease with preexisting CVD or subsequent cardiovascular damage is featured by a significant influx of immune cells and proinflammatory molecules, suggesting that ACE may also participate in COVID-19 induced cardiovascular injury, because COVID-19 disease basically triggers an overactive pathologic immune response. Hopefully, the ACE inhibition and manipulation of those associated bioactive signals could supplement the current medicinal management of various CVD and bring greater benefit to patients' cardiovascular health.
Collapse
Affiliation(s)
- Xinyi Li
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Huasheng Ding
- Department of Emergency, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Gaoke Feng
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Yan Huang
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China.
| |
Collapse
|
7
|
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
|
8
|
Hao XM, Liu Y, Hailaiti D, Gong Y, Zhang XD, Yue BN, Liu JP, Wu XL, Yang KZ, Wang J, Liu QG. Mechanisms of inflammation modulation by different immune cells in hypertensive nephropathy. Front Immunol 2024; 15:1333170. [PMID: 38545112 PMCID: PMC10965702 DOI: 10.3389/fimmu.2024.1333170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 02/15/2024] [Indexed: 04/10/2024] Open
Abstract
Hypertensive nephropathy (HTN) is the second leading cause of end-stage renal disease (ESRD) and a chronic inflammatory disease. Persistent hypertension leads to lesions of intrarenal arterioles and arterioles, luminal stenosis, secondary ischemic renal parenchymal damage, and glomerulosclerosis, tubular atrophy, and interstitial fibrosis. Studying the pathogenesis of hypertensive nephropathy is a prerequisite for diagnosis and treatment. The main cause of HTN is poor long-term blood pressure control, but kidney damage is often accompanied by the occurrence of immune inflammation. Some studies have found that the activation of innate immunity, inflammation and acquired immunity is closely related to the pathogenesis of HTN, which can cause damage and dysfunction of target organs. There are more articles on the mechanism of diabetic nephropathy, while there are fewer studies related to immunity in hypertensive nephropathy. This article reviews the mechanisms by which several different immune cells and inflammatory cytokines regulate blood pressure and renal damage in HTN. It mainly focuses on immune cells, cytokines, and chemokines and inhibitors. However, further comprehensive and large-scale studies are needed to determine the role of these markers and provide effective protocols for clinical intervention and treatment.
Collapse
Affiliation(s)
- Xiao-Min Hao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | | | - Yu Gong
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xu-Dong Zhang
- Department of Chinese Medicine, Beijing Jishuitan Hospital, Beijing, China
| | - Bing-Nan Yue
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ji-Peng Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Li Wu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ke-Zhen Yang
- Department of Rehabilitation Medicine, Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qing-Guo Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Zheng X, Lei Y, Cheng XW. Resolvin D1 as a novel target in the management of hypertension. J Hypertens 2024; 42:393-395. [PMID: 38289998 DOI: 10.1097/hjh.0000000000003641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Affiliation(s)
- Xintong Zheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
| | - Yanna Lei
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, PR China
| |
Collapse
|
10
|
Binda M, Moccaldi B, Civieri G, Cuberli A, Doria A, Tona F, Zanatta E. Autoantibodies Targeting G-Protein-Coupled Receptors: Pathogenetic, Clinical and Therapeutic Implications in Systemic Sclerosis. Int J Mol Sci 2024; 25:2299. [PMID: 38396976 PMCID: PMC10889602 DOI: 10.3390/ijms25042299] [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: 01/10/2024] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Systemic sclerosis (SSc) is a multifaceted connective tissue disease whose aetiology remains largely unknown. Autoimmunity is thought to play a pivotal role in the development of the disease, but the direct pathogenic role of SSc-specific autoantibodies remains to be established. The recent discovery of functional antibodies targeting G-protein-coupled receptors (GPCRs), whose presence has been demonstrated in different autoimmune conditions, has shed some light on SSc pathogenesis. These antibodies bind to GPCRs expressed on immune and non-immune cells as their endogenous ligands, exerting either a stimulatory or inhibitory effect on corresponding intracellular pathways. Growing evidence suggests that, in SSc, the presence of anti-GPCRs antibodies correlates with specific clinical manifestations. Autoantibodies targeting endothelin receptor type A (ETAR) and angiotensin type 1 receptor (AT1R) are associated with severe vasculopathic SSc-related manifestations, while anti-C-X-C motif chemokine receptors (CXCR) antibodies seem to be predictive of interstitial lung involvement; anti-muscarinic-3 acetylcholine receptor (M3R) antibodies have been found in patients with severe gastrointestinal involvement and anti-protease-activated receptor 1 (PAR1) antibodies have been detected in patients experiencing scleroderma renal crisis. This review aims to clarify the potential pathogenetic significance of GPCR-targeting autoantibodies in SSc, focusing on their associations with the different clinical manifestations of scleroderma. An extensive examination of functional autoimmunity targeting GPCRs might provide valuable insights into the underlying pathogenetic mechanisms of SSc, thus enabling the development of novel therapeutic strategies tailored to target GPCR-mediated pathways.
Collapse
Affiliation(s)
- Marco Binda
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, 35128 Padova, Italy; (M.B.)
| | - Beatrice Moccaldi
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, 35128 Padova, Italy; (M.B.)
| | - Giovanni Civieri
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Anna Cuberli
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, 35128 Padova, Italy; (M.B.)
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, 35128 Padova, Italy; (M.B.)
| | - Francesco Tona
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Elisabetta Zanatta
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, 35128 Padova, Italy; (M.B.)
| |
Collapse
|
11
|
Yang Z, Mi X, Li Q, Chen L, Zeng Y, Du P, Liu L, Liu S, Zeng C, Zhang Q, Zhou Y, Xiong Y, Li N, Ze Q, Chen J, He Y. Pharmacokinetic and Bioequivalence Study of Lisinopril/Hydrochlorothiazide Tablet Under Fasting and Postprandial Conditions in Healthy Chinese Subjects. Clin Pharmacol Drug Dev 2024; 13:160-167. [PMID: 37718674 DOI: 10.1002/cpdd.1329] [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: 07/05/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023]
Abstract
The objective of this research was to evaluate and compare the pharmacokinetic profiles and safety of lisinopril/hydrochlorothiazide (10 mg/12.5 mg) tablets in the test and reference formulations administered to participants in both fasting and postprandial states and to evaluate the bioequivalence of the 2 products in healthy Chinese volunteers. This study employed a single-center, randomized, open-label, single-dose dosing trial involving a cumulative 96 healthy adult participants (60 in the fasting group and 36 in the postprandial group). Each group comprised 2 sequence sets, and a 2-week washout period was implemented. There were no statistically significant differences in time to maximum concentration and terminal elimination half-life between the test and control groups under fasting and postprandial conditions (P > .05), and the 90% CIs for area under the plasma concentration-time curve and maximum plasma concentration were within the bioequivalence range of 80%-125%. Pharmacokinetic results indicate a large food effect for lisinopril, meaning that there is a loss of approximately 20%-25% of systemic exposure from fasting to postprandial administration for both preparations. The study demonstrated that a single oral dose of generic lisinopril/hydrochlorothiazide is bioequivalent to the reference product and well tolerated, with no significant adverse events observed, and that both products are similarly safe in a cohort of healthy Chinese male and female participants, following administration under fasting and postprandial conditions.
Collapse
Affiliation(s)
- Zhuan Yang
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Xiaolan Mi
- Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, 300410, China
| | - Qin Li
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lu Chen
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yan Zeng
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Peng Du
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lin Liu
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shijing Liu
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chen Zeng
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qian Zhang
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yan Zhou
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yun Xiong
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Na Li
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qiuyuan Ze
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jiyu Chen
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yan He
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| |
Collapse
|
12
|
Martínez-Casales M, Hernanz R, González-Carnicero Z, Barrús MT, Martín A, Briones AM, Michalska P, León R, Pinilla E, Simonsen U, Alonso MJ. The Melatonin Derivative ITH13001 Prevents Hypertension and Cardiovascular Alterations in Angiotensin II-Infused Mice. J Pharmacol Exp Ther 2024; 388:670-687. [PMID: 38129126 DOI: 10.1124/jpet.123.001586] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 12/23/2023] Open
Abstract
Inflammatory mechanisms and oxidative stress seem to contribute to the pathogenesis of hypertension. ITH13001 is a melatonin-phenyl-acrylate hybrid that moderately induces the antioxidant transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) and has a potent oxidant scavenging effect compared with other derivatives of its family. Here we investigated the effect of ITH13001 on hypertension and the associated cardiovascular alterations. Angiotensin II (AngII)-infused mice were treated with ITH13001 (1 mg/kg per day, i.p.) for 2 weeks. The ITH13001 treatment prevented: 1) the development of hypertension, cardiac hypertrophy, and increased collagen and B-type natriuretic peptide (Bnp) expression in the heart; 2) the reduction of elasticity, incremental distensibility, fenestrae area, intraluminal diameter, and endothelial cell number in mesenteric resistance arteries (MRA); 3) the endothelial dysfunction in aorta and MRA; 4) the plasma and cardiovascular oxidative stress and the reduced aortic nitric oxide (NO) bioavailability; 5) the increased cardiac levels of the cytokines interleukin (IL)-1β, IL-6, and C-C motif chemokine ligand 2 (Ccl2), the T cell marker cluster of differentiation 3 (Cd3), the inflammasome NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3), the proinflammatory enzymes inducible nitric oxide synthase (iNOS) and COX-2, the toll-like receptor 4 (TLR4) adapter protein myeloid differentiation primary response 88 (MyD88), and the nuclear factor kappa B (NF-κB) subunit p65; 6) the greater aortic expression of the cytokines tumor necrosis factor alpha (Tnf-α), Ccl2 and IL-6, Cd3, iNOS, MyD88, and NLRP3. Although ITH13001 increased nuclear Nrf2 levels and heme oxygenase 1 (HO-1) expression in vascular smooth muscle cells, both cardiac and vascular Nrf2, Ho-1, and NADPH quinone dehydrogenase 1 (Nqo1) levels remained unmodified irrespective of AngII infusion. Summarizing, ITH13001 improved hypertension-associated cardiovascular alterations independently of Nrf2 pathway activation, likely due to its direct antioxidant and anti-inflammatory properties. Therefore, ITH13001 could be a useful therapeutic strategy in patients with resistant hypertension. SIGNIFICANCE STATEMENT: Despite the existing therapeutic arsenal, only half of the patients treated for hypertension have adequately controlled blood pressure; therefore, the search for new compounds to control this pathology and the associated damage to end-target organs (cerebral, cardiac, vascular, renal) is of particular interest. The present study demonstrates that a new melatonin derivative, ITH13001, prevents hypertension development and the associated cardiovascular alterations due to its antioxidant and anti-inflammatory properties, making this compound a potential candidate for treatment of resistant hypertensive patients.
Collapse
Affiliation(s)
- Marta Martínez-Casales
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain (M.M.-C., R.H., Z.G.-C, M.T.B., A.M., M.J.A.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (R.H., A.M., A.M.B., M.J.A.); Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain (A.M.B.); Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK (P.M.); Instituto de Química Médica (IQM-CSIC), Madrid, Spain (R.L.); and Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of Health, Aarhus University, Aarhus, Denmark (E.P., U.S.)
| | - Raquel Hernanz
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain (M.M.-C., R.H., Z.G.-C, M.T.B., A.M., M.J.A.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (R.H., A.M., A.M.B., M.J.A.); Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain (A.M.B.); Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK (P.M.); Instituto de Química Médica (IQM-CSIC), Madrid, Spain (R.L.); and Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of Health, Aarhus University, Aarhus, Denmark (E.P., U.S.)
| | - Zoe González-Carnicero
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain (M.M.-C., R.H., Z.G.-C, M.T.B., A.M., M.J.A.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (R.H., A.M., A.M.B., M.J.A.); Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain (A.M.B.); Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK (P.M.); Instituto de Química Médica (IQM-CSIC), Madrid, Spain (R.L.); and Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of Health, Aarhus University, Aarhus, Denmark (E.P., U.S.)
| | - María T Barrús
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain (M.M.-C., R.H., Z.G.-C, M.T.B., A.M., M.J.A.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (R.H., A.M., A.M.B., M.J.A.); Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain (A.M.B.); Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK (P.M.); Instituto de Química Médica (IQM-CSIC), Madrid, Spain (R.L.); and Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of Health, Aarhus University, Aarhus, Denmark (E.P., U.S.)
| | - Angela Martín
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain (M.M.-C., R.H., Z.G.-C, M.T.B., A.M., M.J.A.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (R.H., A.M., A.M.B., M.J.A.); Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain (A.M.B.); Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK (P.M.); Instituto de Química Médica (IQM-CSIC), Madrid, Spain (R.L.); and Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of Health, Aarhus University, Aarhus, Denmark (E.P., U.S.)
| | - Ana M Briones
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain (M.M.-C., R.H., Z.G.-C, M.T.B., A.M., M.J.A.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (R.H., A.M., A.M.B., M.J.A.); Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain (A.M.B.); Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK (P.M.); Instituto de Química Médica (IQM-CSIC), Madrid, Spain (R.L.); and Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of Health, Aarhus University, Aarhus, Denmark (E.P., U.S.)
| | - Patrycja Michalska
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain (M.M.-C., R.H., Z.G.-C, M.T.B., A.M., M.J.A.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (R.H., A.M., A.M.B., M.J.A.); Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain (A.M.B.); Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK (P.M.); Instituto de Química Médica (IQM-CSIC), Madrid, Spain (R.L.); and Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of Health, Aarhus University, Aarhus, Denmark (E.P., U.S.)
| | - Rafael León
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain (M.M.-C., R.H., Z.G.-C, M.T.B., A.M., M.J.A.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (R.H., A.M., A.M.B., M.J.A.); Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain (A.M.B.); Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK (P.M.); Instituto de Química Médica (IQM-CSIC), Madrid, Spain (R.L.); and Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of Health, Aarhus University, Aarhus, Denmark (E.P., U.S.)
| | - Estefano Pinilla
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain (M.M.-C., R.H., Z.G.-C, M.T.B., A.M., M.J.A.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (R.H., A.M., A.M.B., M.J.A.); Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain (A.M.B.); Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK (P.M.); Instituto de Química Médica (IQM-CSIC), Madrid, Spain (R.L.); and Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of Health, Aarhus University, Aarhus, Denmark (E.P., U.S.)
| | - Ulf Simonsen
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain (M.M.-C., R.H., Z.G.-C, M.T.B., A.M., M.J.A.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (R.H., A.M., A.M.B., M.J.A.); Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain (A.M.B.); Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK (P.M.); Instituto de Química Médica (IQM-CSIC), Madrid, Spain (R.L.); and Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of Health, Aarhus University, Aarhus, Denmark (E.P., U.S.)
| | - María J Alonso
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain (M.M.-C., R.H., Z.G.-C, M.T.B., A.M., M.J.A.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (R.H., A.M., A.M.B., M.J.A.); Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain (A.M.B.); Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK (P.M.); Instituto de Química Médica (IQM-CSIC), Madrid, Spain (R.L.); and Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of Health, Aarhus University, Aarhus, Denmark (E.P., U.S.)
| |
Collapse
|
13
|
Bode M, Herrnstadt GR, Dreher L, Ehnert N, Kirkerup P, Lindenmeyer MT, Meyer-Schwesinger CF, Ehmke H, Köhl J, Huber TB, Krebs CF, Steinmetz OM, Wiech T, Wenzel UO. Deficiency of Complement C3a and C5a receptors Does Not Prevent Angiotensin II-Induced Hypertension and Hypertensive End-Organ Damage. Hypertension 2024; 81:138-150. [PMID: 37909169 DOI: 10.1161/hypertensionaha.123.21599] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/12/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND Complement may drive the pathology of hypertension through effects on innate and adaptive immune responses. Recently an injurious role for the anaphylatoxin receptors C3aR (complement component 3a receptor) and C5aR1 (complement component 5a receptor) in the development of hypertension was shown through downregulation of Foxp3+ (forkhead box protein 3) regulatory T cells. Here, we deepen our understanding of the therapeutic potential of targeting both receptors in hypertension. METHODS Data from the European Renal cDNA Bank, single cell sequencing and immunohistochemistry were examined in hypertensive patients. The effect of C3aR or C3aR/C5aR1 double deficiency was assessed in two models of Ang II (angiotensin II)-induced hypertension in knockout mice. RESULTS We found increased expression of C3aR, C5aR1 and Foxp3 cells in kidney biopsies of patients with hypertensive nephropathy. Expression of both receptors was mainly found in myeloid cells. No differences in blood pressure, renal injury (albuminuria, glomerular filtration rate, glomerular and tubulointerstitial injury, inflammation) or cardiac injury (cardiac fibrosis, heart weight, gene expression) between control and mutant mice was discerned in C3aR-/- as well as C3aR/C5aR1-/- double knockout mice. The number of renal Tregs was not decreased in Ang II as well as in DOCA salt induced hypertension. CONCLUSIONS Hypertensive nephropathy in mice and men is characterized by an increase of renal regulatory T cells and enhanced expression of anaphylatoxin receptors. Our investigations do not corroborate a role for C3aR/C5aR1 axis in Ang II-induced hypertension hence challenging the concept of anaphylatoxin receptor targeting in the treatment of hypertensive disease.
Collapse
Affiliation(s)
- Marlies Bode
- III. Department of Medicine (M.B., G.R.H., L.D., N.E., P.K., M.T.L., T.B.H., C.F.K., O.M.S., U.O.W.), University Hospital Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH) (M.B., G.R.H., M.T.L., C.F.M.-S., T.B.H., C.F.K., O.M.S., T.W., U.O.W.)
| | - Georg R Herrnstadt
- III. Department of Medicine (M.B., G.R.H., L.D., N.E., P.K., M.T.L., T.B.H., C.F.K., O.M.S., U.O.W.), University Hospital Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH) (M.B., G.R.H., M.T.L., C.F.M.-S., T.B.H., C.F.K., O.M.S., T.W., U.O.W.)
| | - Leonie Dreher
- III. Department of Medicine (M.B., G.R.H., L.D., N.E., P.K., M.T.L., T.B.H., C.F.K., O.M.S., U.O.W.), University Hospital Hamburg-Eppendorf
| | - Nicolas Ehnert
- III. Department of Medicine (M.B., G.R.H., L.D., N.E., P.K., M.T.L., T.B.H., C.F.K., O.M.S., U.O.W.), University Hospital Hamburg-Eppendorf
| | - Pia Kirkerup
- III. Department of Medicine (M.B., G.R.H., L.D., N.E., P.K., M.T.L., T.B.H., C.F.K., O.M.S., U.O.W.), University Hospital Hamburg-Eppendorf
| | - Maja T Lindenmeyer
- III. Department of Medicine (M.B., G.R.H., L.D., N.E., P.K., M.T.L., T.B.H., C.F.K., O.M.S., U.O.W.), University Hospital Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH) (M.B., G.R.H., M.T.L., C.F.M.-S., T.B.H., C.F.K., O.M.S., T.W., U.O.W.)
| | - Catherine F Meyer-Schwesinger
- Department of Cellular and Integrative Physiology (C.M.-S., H.E.), University Hospital Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH) (M.B., G.R.H., M.T.L., C.F.M.-S., T.B.H., C.F.K., O.M.S., T.W., U.O.W.)
| | - Heimo Ehmke
- Department of Cellular and Integrative Physiology (C.M.-S., H.E.), University Hospital Hamburg-Eppendorf
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, Lübeck, Germany (J.K.)
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, OH (J.K.)
| | - Tobias B Huber
- III. Department of Medicine (M.B., G.R.H., L.D., N.E., P.K., M.T.L., T.B.H., C.F.K., O.M.S., U.O.W.), University Hospital Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH) (M.B., G.R.H., M.T.L., C.F.M.-S., T.B.H., C.F.K., O.M.S., T.W., U.O.W.)
| | - Christian F Krebs
- III. Department of Medicine (M.B., G.R.H., L.D., N.E., P.K., M.T.L., T.B.H., C.F.K., O.M.S., U.O.W.), University Hospital Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH) (M.B., G.R.H., M.T.L., C.F.M.-S., T.B.H., C.F.K., O.M.S., T.W., U.O.W.)
| | - Oliver M Steinmetz
- III. Department of Medicine (M.B., G.R.H., L.D., N.E., P.K., M.T.L., T.B.H., C.F.K., O.M.S., U.O.W.), University Hospital Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH) (M.B., G.R.H., M.T.L., C.F.M.-S., T.B.H., C.F.K., O.M.S., T.W., U.O.W.)
| | - Thorsten Wiech
- Department of Pathology, Section of Nephropathology (T.W.), University Hospital Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH) (M.B., G.R.H., M.T.L., C.F.M.-S., T.B.H., C.F.K., O.M.S., T.W., U.O.W.)
| | - Ulrich O Wenzel
- III. Department of Medicine (M.B., G.R.H., L.D., N.E., P.K., M.T.L., T.B.H., C.F.K., O.M.S., U.O.W.), University Hospital Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH) (M.B., G.R.H., M.T.L., C.F.M.-S., T.B.H., C.F.K., O.M.S., T.W., U.O.W.)
| |
Collapse
|
14
|
Schmidt-Lauber C, Alba Schmidt E, Hänzelmann S, Petersen EL, Behrendt CA, Twerenbold R, Blankenberg S, Huber TB, Wenzel UO. Increased blood pressure after nonsevere COVID-19. J Hypertens 2023; 41:1721-1729. [PMID: 37682048 PMCID: PMC10552808 DOI: 10.1097/hjh.0000000000003522] [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: 03/05/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Various sequelae have been described after nonsevere coronavirus disease 2019 (COVID-19), but knowledge on postacute effects on blood pressure is limited. METHODS This is a cross-sectional analysis of blood pressure profiles in individuals after nonsevere COVID-19 compared with matched population-based individuals without prior COVID-19. Data were derived from the ongoing and prospective Hamburg City Health Study, a population-based study in Hamburg, Germany, and its associated COVID-19 program, which included individuals at least 4 months after COVID-19. Matching was performed by age, sex, education, and preexisting hypertension in a 1 : 4 ratio. RESULTS Four hundred and thirty-two individuals after COVID-19 (mean age 56.1 years) were matched to 1728 controls without prior COVID-19 (56.2 years). About 92.8% of COVID-19 courses were mild or moderate, only 7.2% were hospitalized, and no individual had been treated on an intensive care unit. Even after adjustment for relevant competing risk factors, DBP [+4.7 mmHg, 95% confidence interval (95% CI) 3.97-5.7, P < 0.001] was significantly higher in individuals after COVID-19. For SBP, a trend towards increased values was observed (+1.4 mmHg, 95% CI -0.4 to 3.2, P = 0.120). Hypertensive blood pressures at least 130/80 mmHg (according to the ACC/AHA guideline) and at least 140/90 mmHg (ESC/ESH guideline) occurred significantly more often in individuals after COVID-19 than matched controls (odds ratio 2.0, 95% CI 1.5-2.7, P < 0.001 and odds ratio 1.6, 95% CI 1.3-2.0, P < 0.001, respectively), mainly driven by changes in DBP. CONCLUSION Blood pressure is higher in individuals after nonsevere COVID-19 compared with uninfected individuals suggesting a significant hypertensive sequela.
Collapse
Affiliation(s)
- Christian Schmidt-Lauber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg
- Research Center On Rare Kidney Diseases (RECORD), University Hospital Erlangen, Erlangen
| | - Elisa Alba Schmidt
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg
| | - Sonja Hänzelmann
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf
| | - Elina L. Petersen
- Department of Cardiology, University Heart and Vascular Center, Hamburg
| | | | - Raphael Twerenbold
- Department of Cardiology, University Heart and Vascular Center, Hamburg
- University Center of Cardiovascular Science, University Medical Center Hamburg-Eppendorf, Hamburg
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg–Kiel–Lübeck, Germany
| | - Stefan Blankenberg
- Department of Cardiology, University Heart and Vascular Center, Hamburg
- University Center of Cardiovascular Science, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg
| | - Ulrich O. Wenzel
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg
| |
Collapse
|
15
|
Bode M, Diemer JN, Luu TV, Ehnert N, Teigeler T, Wiech T, Lindenmeyer MT, Herrnstadt GR, Bülow J, Huber TB, Tomas NM, Wenzel UO. Complement component C3 as a new target to lower albuminuria in hypertensive kidney disease. Br J Pharmacol 2023; 180:2412-2435. [PMID: 37076314 DOI: 10.1111/bph.16097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND AND PURPOSE Complement activation may drive hypertension through its effects on immunity and tissue integrity. EXPERIMENTAL APPROACH We examined expression of C3, the central protein of the complement cascade, in hypertension. KEY RESULTS Increased C3 expression was found in kidney biopsies and micro-dissected glomeruli of patients with hypertensive nephropathy. Renal single cell RNA sequence data from normotensive and hypertensive patients confirmed expression of C3 in different cellular compartments of the kidney. In angiotensin II (Ang II) induced hypertension renal C3 expression was up-regulated. C3-/- mice revealed a significant lower albuminuria in the early phase of hypertension. However, no difference was found for blood pressure, renal injury (histology, glomerular filtration rate, inflammation) and cardiac injury (fibrosis, weight, gene expression) between C3-/- and wildtype mice after Ang II infusion. Also, in deoxycorticosterone acetate (DOCA) salt hypertension, a significantly lower albuminuria was found in the first weeks of hypertension in C3 deficient mice but no significant difference in renal and cardiac injury. Down-regulation of C3 by C3 targeting GalNAc (n-acetylgalactosamine) small interfering RNA (siRNA) conjugate decreased C3 in the liver by 96% and lowered albuminuria in the early phase but showed no effect on blood pressure and end-organ damage. Inhibition of complement C5 by siRNA showed no effect on albuminuria. CONCLUSION AND IMPLICATIONS Increased C3 expression is found in the kidneys of hypertensive mice and men. Genetic and therapeutic knockdown of C3 improved albuminuria in the early phase of hypertension but did not ameliorate arterial blood pressure nor renal and cardiac injury.
Collapse
Affiliation(s)
- Marlies Bode
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Niklas Diemer
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - The Vinh Luu
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Nikolas Ehnert
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Teresa Teigeler
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Wiech
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute for Pathology, Section Nephropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maja T Lindenmeyer
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Georg R Herrnstadt
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Jasmin Bülow
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola M Tomas
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich O Wenzel
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
16
|
Popescu D, Rezus E, Badescu MC, Dima N, Seritean Isac PN, Dragoi IT, Rezus C. Cardiovascular Risk Assessment in Rheumatoid Arthritis: Accelerated Atherosclerosis, New Biomarkers, and the Effects of Biological Therapy. Life (Basel) 2023; 13:life13020319. [PMID: 36836675 PMCID: PMC9965162 DOI: 10.3390/life13020319] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Rheumatoid arthritis (RA), one of the most common of the chronic inflammatory autoimmune diseases (CIADs), is recognized as an independent cardiovascular risk factor. Traditional risk factors such as smoking, arterial hypertension, dyslipidemia, insulin resistance, and obesity are frequently found in RA. Given the increased risk of mortality and morbidity associated with cardiovascular disease (CVD) in RA patients, screening for risk factors is important. Moreover, there is a need to identify potential predictors of subclinical atherosclerosis. Recent studies have shown that markers such as serum homocysteine, asymmetric dimethylarginine, or carotid intima-media thickness (cIMT) are correlated with cardiovascular risk. Although RA presents a cardiovascular risk comparable to that of diabetes, it is not managed as well in terms of acute cardiovascular events. The introduction of biological therapy has opened new perspectives in the understanding of this pathology, confirming the involvement and importance of the inflammatory markers, cytokines, and the immune system. In addition to effects in inducing remission and slowing disease progression, most biologics have demonstrated efficacy in reducing the risk of major cardiovascular events. Some studies have also been conducted in patients without RA, with similar results. However, early detection of atherosclerosis and the use of targeted therapies are the cornerstone for reducing cardiovascular risk in RA patients.
Collapse
Affiliation(s)
- Diana Popescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology and Physiotherapy, “Grigore. T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Rheumatology Clinic, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
- Correspondence: (E.R.); (M.C.B.)
| | - Minerva Codruta Badescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
- Correspondence: (E.R.); (M.C.B.)
| | - Nicoleta Dima
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Petronela Nicoleta Seritean Isac
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Ioan-Teodor Dragoi
- Department of Rheumatology and Physiotherapy, “Grigore. T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Rheumatology Clinic, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| |
Collapse
|
17
|
Kostoff RN, Briggs MB, Kanduc D, Dewanjee S, Kandimalla R, Shoenfeld Y, Porter AL, Tsatsakis A. Modifiable contributing factors to COVID-19: A comprehensive review. Food Chem Toxicol 2023; 171:113511. [PMID: 36450305 PMCID: PMC9701571 DOI: 10.1016/j.fct.2022.113511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/29/2022]
Abstract
The devastating complications of coronavirus disease 2019 (COVID-19) result from an individual's dysfunctional immune response following the initial severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Multiple toxic stressors and behaviors contribute to underlying immune system dysfunction. SARS-CoV-2 exploits the dysfunctional immune system to trigger a chain of events ultimately leading to COVID-19. The current study identifies eighty immune system dysfunction-enabling toxic stressors and behaviors (hereafter called modifiable contributing factors (CFs)) that also link directly to COVID-19. Each CF is assigned to one of the five categories in the CF taxonomy shown in Section 3.3.: Lifestyle (e.g., diet, substance abuse); Iatrogenic (e.g., drugs, surgery); Biotoxins (e.g., micro-organisms, mycotoxins); Occupational/Environmental (e.g., heavy metals, pesticides); Psychosocial/Socioeconomic (e.g., chronic stress, lower education). The current study shows how each modifiable factor contributes to decreased immune system capability, increased inflammation and coagulation, and increased neural damage and neurodegeneration. It is unclear how real progress can be made in combatting COVID-19 and other similar diseases caused by viral variants without addressing and eliminating these modifiable CFs.
Collapse
Affiliation(s)
- Ronald Neil Kostoff
- Independent Consultant, Gainesville, VA, 20155, USA,Corresponding author. Independent Consultant, 13500 Tallyrand Way, Gainesville, VA, 20155, USA
| | | | - Darja Kanduc
- Dept. of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Via Orabona 4, Bari, 70125, Italy
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, 500007, Telangana, India
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, 5265601, Israel
| | - Alan L. Porter
- School of Public Policy, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003, Heraklion, Greece
| |
Collapse
|
18
|
Lymphocyte to monocyte ratio and blood pressure variability in childhood hypertension-a pilot study. Pediatr Res 2023; 93:137-142. [PMID: 35379928 DOI: 10.1038/s41390-022-02056-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/23/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS The investigation of inflammatory background of hypertension (HTN) concentrates mainly on patients with primary HTN. The aim of the study was to analyze the role of new parameters of inflammation-lymphocyte to monocyte ratio (LMR), neutrophil to lymphocyte ratio (NLR), and platelet to lymphocyte ratio (PLR), in the population of children with primary (pHTN) and secondary renal hypertension (rHTN). MATERIAL AND METHODS The study group consisted of 70 children with pHTN, 46 patients with rHTN, and 30 age-matched normotensive controls. The retrospective analysis focused on the evaluation of LMR, NLR, and PLR values in relation to blood pressure (BP) parameters from in-office and ambulatory BP monitoring measurements. Twenty-four hours, daytime, and nighttime periods were evaluated. Blood pressure variability (BPV) was defined by standard deviation and coefficient of variation of analyzed values. RESULTS LMR and NLR values in HTN patients differed significantly vs. controls. Dippers with pHTN demonstrated significant correlations between LMR, NLR, PLR, and markers of BPV, in 24 h and daytime diastolic BP and mean arterial pressure. In dippers with rHTN such correlations concerned only LMR. CONCLUSIONS LMR may become a promising marker of BPV, useful in children with primary and secondary hypertension. IMPACT Lymphocyte to monocyte ratio is a novel marker of blood pressure variability, connected to target-organ damage, in children with primary and secondary renal hypertension. Our study analyzes for the first time the connections between blood cell count-driven inflammatory markers (lymphocyte to monocyte, neutrophil to lymphocyte, and platelet to lymphocyte ratios) and parameters of blood pressure variability, and compares those ratios in children with primary and secondary hypertension. The increasing incidence of hypertension among children urges the search for simple methods of assessment of its complications. LMR may be of added value in the analysis of the inflammatory background of hypertension.
Collapse
|
19
|
Liao L, Zhang L, Chen H, Teng D, Xu B, Gong L, Zhong L, Wang C, Dong H, Jia W, Yang J, Shi Z. Identification of Key Genes from the Visceral Adipose Tissues of Overweight/Obese Adults with Hypertension through Transcriptome Sequencing. Cytogenet Genome Res 2022; 162:541-559. [PMID: 36521430 PMCID: PMC10534961 DOI: 10.1159/000528702] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/12/2022] [Indexed: 08/31/2023] Open
Abstract
Overweight and obese (OW/OB) adults are at increased risk of hypertension due to visceral adipose tissue (VAT) inflammation. In this study, we explored gene level differences in the VAT of hypertensive and normotensive OW/OB patients. VAT samples obtained from six OW/OB adults (three hypertensive, three normotensive) were subjected to transcriptome sequencing analysis. Gene set enrichment analysis was conducted for all gene expression data to identify differentially expressed genes (DEGs) with |log2 (fold change)| ≥ 1 and q < 0.05. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional enrichment analyses were performed on the DEGs, and hub genes were identified by constructing a protein-protein interaction (PPI) network. The proposed hub genes were validated using quantitative real-time PCR in ten other samples from five hypertensive and five normotensive patients. In addition, we performed ROC analysis and Spearman correlation analysis. A total of 84 DEGs were identified between VAT samples from OW/OB patients with and without hypertension, among which 21 were significantly upregulated and 63 were significantly downregulated. Bioinformatics analysis revealed that spleen function was related to hypertension in OW/OB adults. Meanwhile, PPI network analysis identified the following top 10 hub genes: CD79A, CR2, SELL, CD22, IL7R, CCR7, TNFRSF13C, CXCR4, POU2AF1, and JAK3. Through qPCR verification, we found that CXCR4, CD22, and IL7R were statistically significant. qPCR verification suggested that RELA was statistically significant. However, qPCR verification indicated that NFKB1 and KLF2 were not statistically significant. These hub genes were mainly regulated by the transcription factor RELA. The AUC of ROC analysis for CXCR4, IL7R, and CD22 was 0.92. What is more, VAT CXCR4 and CD22 were positively related to RELA relative expression levels. Taken together, our research demonstrates that CXCR4, IL7R, and CD22 related to VAT in hypertensive OW/OB adults could serve as future therapeutic targets.
Collapse
Affiliation(s)
- Lanlan Liao
- The Second Clinical Medical College, Binzhou Medical University, Yantai, China
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Lihui Zhang
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai, China
- Medical College, Qingdao University, Qingdao, China
| | - Hongping Chen
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai, China
- Medical College, Qingdao University, Qingdao, China
| | - Da Teng
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai, China
- Medical College, Qingdao University, Qingdao, China
| | - Bowen Xu
- The Second Clinical Medical College, Binzhou Medical University, Yantai, China
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Lei Gong
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Lin Zhong
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Chunxiao Wang
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Haibin Dong
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Wenjuan Jia
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Jun Yang
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Zhen Shi
- Basic Medical College, Binzhou Medical University, Yantai, China
| |
Collapse
|
20
|
Long non-coding RNAs as biomarkers and therapeutic targets for ischemic stroke. Noncoding RNA Res 2022; 7:226-232. [PMID: 36187570 PMCID: PMC9508273 DOI: 10.1016/j.ncrna.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 11/30/2022] Open
Abstract
Background The problem of ischemic stroke (IS) has become increasingly important in recent years, as it ranks first in the structure of disability and mortality, crowding out other vascular diseases. In this regard, the study of this pathology and the search for new therapeutic and diagnostic tools remains an urgent problem of modern medical science and practice. Long non-coding RNAs (lncRNAs)-based therapeutics and diagnostic tools offer a very attractive area of study. Therefore, this systematic review aims at summarizing current knowledge on promising lncRNAs as biomarkers and therapeutic targets for IS exploring original articles and literature reviews on in vivo, in vitro and ex vivo experiments. Methods The current systematic review was performed according to PRISMA guidelines. PubMed, MEDLINE and Google Scholar databases were comprehensively explored to perform the article search. Results 34 eligible studies were included and analyzed: 25 focused on lncRNAs-based therapeutics and 9 on lncRNAs-based diagnosis. We found 31 different lncRNAs tested as potential therapeutic and diagnostic molecules in cells and animal model experiments. Among all founded lncRNA-based therapeutics and non-invasive diagnostic tools, nuclear enriched abundant transcript 1 (NEAT1) emerged to be the most investigated and proposed as a potential molecule for IS diagnosis and treatment. Conclusions Our analysis provides a snapshot of the current scenario regarding the lncRNAs as therapeutic molecules and biomarkers in IS. Different lncRNAs are differently expressed in IS, and some of them can be further evaluated as therapeutic targets and biomarkers for early diagnosis and prognosis or treatment response. However, despite many efforts, none of the selected studies go beyond preclinical studies, and their translation into clinical practice seems to be very premature.
Collapse
|
21
|
Liska DJ, Dioum E, Chu Y, Mah E. Narrative Review on the Effects of Oat and Sprouted Oat Components on Blood Pressure. Nutrients 2022; 14:nu14224772. [PMID: 36432463 PMCID: PMC9698489 DOI: 10.3390/nu14224772] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Hypertension (HTN) is a major risk factor for cardiovascular disease (CVD) and cognitive decline. Elevations in blood pressure (BP) leading to HTN can be found in young adults with increased prevalence as people age. Oats are known to decrease CVD risk via an established effect of β-glucan on the attenuation of blood cholesterol. Many past studies on CVD and oats have also reported a decrease in BP; however, a thorough assessment of oats and BP has not been conducted. Moreover, oats deliver several beneficial dietary components with putative beneficial effects on BP or endothelial function, such as β-glucan, γ-amino butyric acid (GABA), and phytochemicals such as avenanthramides. We conducted a comprehensive search for systematic reviews, meta-analyses, and clinical intervention studies on oats and BP and identified 18 randomized controlled trials (RCTs) and three meta-analyses that supported the role of oats in decreasing BP. Emerging data also suggest oat consumption may reduce the use of anti-hypertensive medications. The majority of these studies utilized whole oats or oat bran, which include a vast array of oat bioactives. Therefore, we also extensively reviewed the literature on these bioactives and their putative effect on BP-relevant mechanisms. The data suggest several oat components, such as GABA, as well as the delivery of high-quality plant protein and fermentable prebiotic fiber, may contribute to the anti-HTN effect of oats. In particular, GABA is enhanced in oat sprouts, which suggests this food may be particularly beneficial for healthy BP management.
Collapse
Affiliation(s)
| | - ElHadji Dioum
- PepsiCo R&D Health & Nutrition Sciences, Chicago, IL 60607, USA
| | - Yifang Chu
- PepsiCo R&D Health & Nutrition Sciences, Chicago, IL 60607, USA
| | - Eunice Mah
- Biofortis Research, Addison, IL 60101, USA
- Correspondence:
| |
Collapse
|
22
|
Könnecke H, Schnabel RB, Walther C, Lamprecht R, Heydecke G, Seedorf U, Jagodzinski A, Borof K, Zeller T, Beikler T, Smeets R, Gosau M, Behrendt CA, Wenzel U, Börschel CS, Karakas M, Blankenberg S, Aarabi G. Cross-sectional study on the association of periodontitis with arterial hypertension in the Hamburg City Health Study. Eur J Med Res 2022; 27:181. [PMID: 36114562 PMCID: PMC9479239 DOI: 10.1186/s40001-022-00811-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 09/05/2022] [Indexed: 09/05/2024] Open
Abstract
AIM Aim of this study was to investigate the association between periodontitis and arterial hypertension, both of which show correlations with classical cardiovascular risk factors and inflammatory activity. MATERIALS AND METHODS A cross-sectional analysis of data from a large population-based health survey (the Hamburg City Health Study, HCHS) including 5934 participants with complete periodontal examination and blood pressure data, of whom 5735 had medical records regarding anti-hypertensive medication, was performed. Probing depths, gingival recessions, bleeding on probing (BOP), dental plaque, and decayed-missing-filled teeth (DMFT) indices were recorded as measures of oral health. Clinical attachment loss (CAL) per tooth was calculated and periodontitis was staged into three groups (no/mild, moderate, severe). Arterial hypertension was diagnosed based on the participants' medication history and systolic and diastolic blood pressure values. Logistic regression models were constructed accounting for a set of potential confounders (age, sex, smoking, body mass index (BMI), diabetes, educational level, alcohol intake) and high sensitivity-C-reactive protein (hsCRP). RESULTS The odds of arterial hypertension increased significantly along with periodontitis severity (OR for severe periodontitis: 2.19; 95% CI 1.85-2.59; p < 0.001; OR for moderate periodontitis: 1.65; 95% CI 1.45-1.87; p < 0.001). Participants with moderate or severe periodontitis also had significantly higher age- and sex-adjusted odds of arterial hypertension, which was slightly weakened when additionally adjusted for BMI, diabetes, smoking, educational level, and alcohol intake (OR for severe PD: 1.28, 95% CI 1.04-1.59, p = 0.02; OR for moderate PD: 1.30, 95% CI 1.11-1.52, p = 0.001). The fraction of participants with undertreated hypertension (untreated and poorly controlled hypertension) was considerably larger in participants with severe periodontitis than in those with no/mild periodontitis (50.1% vs. 37.4% for no/mild periodontitis). CONCLUSIONS The study shows an association between periodontitis and arterial hypertension that is independent of age, sex, diabetes, BMI, smoking, educational level, and alcohol intake. In addition, undertreatment of hypertension was more common in people with severe periodontitis compared with periodontally more healthy people.
Collapse
Affiliation(s)
- Henrieke Könnecke
- Department of Prosthetic Dentistry, Center for Dental and Oral Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251, Hamburg, Germany
| | - Renate B Schnabel
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg, Kiel, Lübeck, Germany
| | - Carolin Walther
- Department of Prosthetic Dentistry, Center for Dental and Oral Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251, Hamburg, Germany
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ragna Lamprecht
- Department of Prosthetic Dentistry, Center for Dental and Oral Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251, Hamburg, Germany
| | - Guido Heydecke
- Department of Prosthetic Dentistry, Center for Dental and Oral Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251, Hamburg, Germany
| | - Udo Seedorf
- Department of Prosthetic Dentistry, Center for Dental and Oral Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251, Hamburg, Germany
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annika Jagodzinski
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
- Epidemiological Study Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Borof
- Epidemiological Study Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Zeller
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg, Kiel, Lübeck, Germany
| | - Thomas Beikler
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Oral and Maxillofacial Surgery, Division of "Regenerative Orofacial Medicine", University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Gosau
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Ulrich Wenzel
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christin S Börschel
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg, Kiel, Lübeck, Germany
| | - Mahir Karakas
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg, Kiel, Lübeck, Germany
| | - Stefan Blankenberg
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg, Kiel, Lübeck, Germany
| | - Ghazal Aarabi
- Department of Prosthetic Dentistry, Center for Dental and Oral Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251, Hamburg, Germany.
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
23
|
Zhang Z, Dalan R, Hu Z, Wang JW, Chew NW, Poh KK, Tan RS, Soong TW, Dai Y, Ye L, Chen X. Reactive Oxygen Species Scavenging Nanomedicine for the Treatment of Ischemic Heart Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202169. [PMID: 35470476 DOI: 10.1002/adma.202202169] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/08/2022] [Indexed: 06/14/2023]
Abstract
Ischemic heart disease (IHD) is the leading cause of disability and mortality worldwide. Reactive oxygen species (ROS) have been shown to play key roles in the progression of diabetes, hypertension, and hypercholesterolemia, which are independent risk factors that lead to atherosclerosis and the development of IHD. Engineered biomaterial-based nanomedicines are under extensive investigation and exploration, serving as smart and multifunctional nanocarriers for synergistic therapeutic effect. Capitalizing on cell/molecule-targeting drug delivery, nanomedicines present enhanced specificity and safety with favorable pharmacokinetics and pharmacodynamics. Herein, the roles of ROS in both IHD and its risk factors are discussed, highlighting cardiovascular medications that have antioxidant properties, and summarizing the advantages, properties, and recent achievements of nanomedicines that have ROS scavenging capacity for the treatment of diabetes, hypertension, hypercholesterolemia, atherosclerosis, ischemia/reperfusion, and myocardial infarction. Finally, the current challenges of nanomedicines for ROS-scavenging treatment of IHD and possible future directions are discussed from a clinical perspective.
Collapse
Affiliation(s)
- Zhan Zhang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 408433, Singapore
| | - Zhenyu Hu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jiong-Wei Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Nicholas Ws Chew
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Kian-Keong Poh
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Ru-San Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, 119609, Singapore
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macao, Taipa, Macau SAR, 999078, China
| | - Lei Ye
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Chemical and Biomolecular Engineering and Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
24
|
Luettges K, Bode M, Diemer JN, Schwanbeck J, Wirth EK, Klopfleisch R, Kappert K, Thiele A, Ritter D, Foryst-Ludwig A, Kolkhof P, Wenzel UO, Kintscher U. Finerenone Reduces Renal RORγt γδ T Cells and Protects against Cardiorenal Damage. Am J Nephrol 2022; 53:552-564. [PMID: 35675794 DOI: 10.1159/000524940] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/27/2022] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Chronic activation of the mineralocorticoid receptor (MR) leads to pathological processes like inflammation and fibrosis during cardiorenal disease. Modulation of immunological processes in the heart or kidney may serve as a mechanistic and therapeutic interface in cardiorenal pathologies. In this study, we investigated anti-inflammatory/-fibrotic and immunological effects of the selective nonsteroidal MR antagonists finerenone (FIN) in the deoxycorticosterone acetate (DOCA)-salt model. METHODS Male C57BL6/J mice were uninephrectomized and received a DOCA pellet implantation (2.4 mg/day) plus 0.9% NaCl in drinking water (DOCA-salt) or received a sham operation and were orally treated with FIN (10 mg/kg/day) or vehicle in a preventive study design. Five weeks after the procedure, blood pressure (BP), urinary albumin/creatinine ratio (UACR), glomerular and tubulointerstitial damage, echocardiographic cardiac function, as well as cardiac/renal inflammatory cell content by FACS analysis were assessed. RESULTS BP was significantly reduced by FIN. FACS analysis revealed a notable immune response due to DOCA-salt exposure. Especially, infiltrating renal RORγt γδ-positive T cells were upregulated, which was significantly ameliorated by FIN treatment. This was accompanied by a significant reduction of UACR in FIN-treated mice. In the heart, FIN reduced DOCA-salt-induced cardiac hypertrophy, cardiac fibrosis and led to an improvement of the global longitudinal strain. Cardiac actions of FIN were not associated with a regulation of cardiac RORγt γδ-positive T cells. DISCUSSION/CONCLUSION The present study shows cardiac and renal protective effects of FIN in a DOCA-salt model. The cardiorenal protection was accompanied by a reduction of renal RORγt γδ T cells. The observed actions of FIN may provide a potential mechanism of its efficacy recently observed in clinical trials.
Collapse
Affiliation(s)
- Katja Luettges
- Cardiovascular Metabolic Renal Research Center, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Marlies Bode
- Department of Medicine III, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Jan Niklas Diemer
- Department of Medicine III, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Juliane Schwanbeck
- Cardiovascular Metabolic Renal Research Center, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Eva K Wirth
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Kai Kappert
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin Humboldt-Universität zu Berlin, Berlin, Germany
| | - Arne Thiele
- Cardiovascular Metabolic Renal Research Center, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Daniel Ritter
- Cardiovascular Metabolic Renal Research Center, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Anna Foryst-Ludwig
- Cardiovascular Metabolic Renal Research Center, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Peter Kolkhof
- Bayer AG, Research & Early Development, Cardiovascular Research, Wuppertal, Germany
| | - Ulrich Otto Wenzel
- Department of Medicine III, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Ulrich Kintscher
- Cardiovascular Metabolic Renal Research Center, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
25
|
Hengel FE, Benitah JP, Wenzel UO. Mosaic theory revised: inflammation and salt play central roles in arterial hypertension. Cell Mol Immunol 2022; 19:561-576. [PMID: 35354938 PMCID: PMC9061754 DOI: 10.1038/s41423-022-00851-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
The mosaic theory of hypertension was advocated by Irvine Page ~80 years ago and suggested that hypertension resulted from the close interactions of different causes. Increasing evidence indicates that hypertension and hypertensive end-organ damage are not only mediated by the proposed mechanisms that result in hemodynamic injury. Inflammation plays an important role in the pathophysiology and contributes to the deleterious consequences of arterial hypertension. Sodium intake is indispensable for normal body function but can be detrimental when it exceeds dietary requirements. Recent data show that sodium levels also modulate the function of monocytes/macrophages, dendritic cells, and different T-cell subsets. Some of these effects are mediated by changes in the microbiome and metabolome due to high-salt intake. The purpose of this review is to propose a revised and extended version of the mosaic theory by summarizing and integrating recent advances in salt, immunity, and hypertension research. Salt and inflammation are placed in the middle of the mosaic because both factors influence each of the remaining pieces.
Collapse
Affiliation(s)
- Felicitas E Hengel
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Jean-Pierre Benitah
- Inserm UMR-S 1180, Faculty of Pharmacy, University Paris Saclay, Gif-sur-Yvette, France
| | - Ulrich O Wenzel
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
26
|
van der Heijden CDCC, Bode M, Riksen NP, Wenzel UO. The role of the mineralocorticoid receptor in immune cells in in cardiovascular disease. Br J Pharmacol 2021; 179:3135-3151. [PMID: 34935128 DOI: 10.1111/bph.15782] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/22/2021] [Accepted: 11/22/2021] [Indexed: 11/29/2022] Open
Abstract
Chronic low-grade inflammation and immune cell activation are important mechanisms in the pathophysiology of cardiovascular disease (CVD). Therefore, targeted immunosuppression is a promising novel therapy to lower cardiovascular risk. In this review, we identify the mineralocorticoid receptor (MR) on immune cells as a potential target to modulate inflammation. The MR is present in almost all cells of the cardiovascular system, including immune cells. Activation of the MR in innate and adaptive immune cells induces inflammation which can contribute to CVD, by inducing endothelial dysfunction and hypertension. Moreover, it accelerates atherosclerotic plaque formation and destabilization and impairs tissue regeneration after ischemic events. Identifying the molecular targets for these non-renal actions of the MR provide promising novel cardiovascular drug targets for mineralocorticoid receptor antagonists (MRAs), which are currently mainly applied in hypertension and heart failure.
Collapse
Affiliation(s)
| | - Marlies Bode
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, GA, Nijmegen, The Netherlands.,Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, GA, The Netherlands
| | - Ulrich O Wenzel
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
27
|
Kountouras J, Papaefthymiou A, Polyzos SA, Deretzi G, Vardaka E, Soteriades ES, Tzitiridou-Chatzopoulou M, Gkolfakis P, Karafyllidou K, Doulberis M. Impact of Helicobacter pylori-Related Metabolic Syndrome Parameters on Arterial Hypertension. Microorganisms 2021; 9:microorganisms9112351. [PMID: 34835476 PMCID: PMC8618184 DOI: 10.3390/microorganisms9112351] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/28/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Arterial hypertension is a risk factor for several pathologies, mainly including cardio-cerebrovascular diseases, which rank as leading causes of morbidity and mortality worldwide. Arterial hypertension also constitutes a fundamental component of the metabolic syndrome. Helicobacter pylori infection is one of the most common types of chronic infection globally and displays a plethora of both gastric and extragastric effects. Among other entities, Helicobacter pylori has been implicated in the pathogenesis of the metabolic syndrome. Within this review, we illustrate the current state-of-the-art evidence, which may link several components of the Helicobacter pylori-related metabolic syndrome, including non-alcoholic fatty liver disease and arterial hypertension. In particular, current knowledge of how Helicobacter pylori exerts its virulence through dietary, inflammatory and metabolic pathways will be discussed. Although there is still no causative link between these entities, the emerging evidence from both basic and clinical research supports the proposal that several components of the Helicobacter pylori infection-related metabolic syndrome present an important risk factor in the development of arterial hypertension. The triad of Helicobacter pylori infection, the metabolic syndrome, and hypertension represents a crucial worldwide health problem on a pandemic scale with high morbidity and mortality, like COVID-19, thereby requiring awareness and appropriate management on a global scale.
Collapse
Affiliation(s)
- Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- Correspondence:
| | - Apostolis Papaefthymiou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- Department of Gastroenterology, University Hospital of Larisa, 41110 Larisa, Greece
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Stergios A. Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Georgia Deretzi
- Multiple Sclerosis Unit, Department of Neurology, Papageorgiou General Hospital, 56403 Thessaloniki, Greece;
| | - Elisabeth Vardaka
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece;
| | - Elpidoforos S. Soteriades
- Healthcare Management Program, School of Economics and Management, Open University of Cyprus, Nicosia 2252, Cyprus;
- Department of Environmental Health, Environmental and Occupational Medicine and Epidemiology (EOME), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, 50100 Kozani, Greece
| | - Paraskevas Gkolfakis
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Erasme University Hospital, 1070 Brussels, Belgium;
- Department of Medical Oncology, Institut Jules Bordet, 1000 Brussels, Belgium
| | - Kyriaki Karafyllidou
- Department of Pediatrics, University Children’s Hospital of Zurich, 8032 Zurich, Switzerland;
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland
| |
Collapse
|
28
|
Boesen EI, Kakalij RM. Autoimmune-mediated renal disease and hypertension. Clin Sci (Lond) 2021; 135:2165-2196. [PMID: 34533582 PMCID: PMC8477620 DOI: 10.1042/cs20200955] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 08/20/2021] [Accepted: 09/06/2021] [Indexed: 12/18/2022]
Abstract
Hypertension is a major risk factor for cardiovascular disease, chronic kidney disease (CKD), and mortality. Troublingly, hypertension is highly prevalent in patients with autoimmune renal disease and hastens renal functional decline. Although progress has been made over the past two decades in understanding the inflammatory contributions to essential hypertension more broadly, the mechanisms active in autoimmune-mediated renal diseases remain grossly understudied. This Review provides an overview of the pathogenesis of each of the major autoimmune diseases affecting the kidney that are associated with hypertension, and describes the current state of knowledge regarding hypertension in these diseases and their management. Specifically, discussion focuses on Systemic Lupus Erythematosus (SLE) and Lupus Nephritis (LN), Immunoglobulin A (IgA) Nephropathy, Idiopathic Membranous Nephropathy (IMN), Anti-Neutrophil Cytoplasmic Antibody (ANCA)-associated glomerulonephritis, and Thrombotic Thrombocytopenic Purpura (TTP). A summary of disease-specific animal models found to exhibit hypertension is also included to highlight opportunities for much needed further investigation of underlying mechanisms and novel therapeutic approaches.
Collapse
Affiliation(s)
- Erika I Boesen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| | - Rahul M Kakalij
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| |
Collapse
|
29
|
Immune-mediated glomerular diseases: new basic concepts and clinical implications. Cell Tissue Res 2021; 385:277-279. [PMID: 34463820 PMCID: PMC8523466 DOI: 10.1007/s00441-021-03509-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 11/18/2022]
|
30
|
Genotypic analysis of the female BPH/5 mouse, a model of superimposed preeclampsia. PLoS One 2021; 16:e0253453. [PMID: 34270549 PMCID: PMC8284809 DOI: 10.1371/journal.pone.0253453] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/04/2021] [Indexed: 11/19/2022] Open
Abstract
Animal models that recapitulate human diseases and disorders are widely used to investigate etiology, diagnosis, and treatment of those conditions in people. Disorders during pregnancy are particularly difficult to explore as interventions in pregnant women are not easily performed. Therefore, models that allow for pre-conception investigations are advantageous for elucidating the mechanisms involved in adverse pregnancy outcomes that are responsible for both maternal and fetal morbidity, such as preeclampsia. The Blood Pressure High (BPH)/5 mouse model has been used extensively to study the pathogenesis of preeclampsia. The female BPH/5 mouse is obese with increased adiposity and borderline hypertension, both of which are exacerbated with pregnancy making it a model of superimposed preeclampsia. Thus, the BPH/5 model shares traits with a large majority of women with pre-existing conditions that predisposes them to preeclampsia. We sought to explore the genome of the BPH/5 female mouse and determine the genetic underpinnings that may contribute to preeclampsia-associated phenotypes in this model. Using a whole genome sequencing approach, we are the first to characterize the genetic mutations in BPH/5 female mice that make it unique from the closely related BPH/2 model and the normotensive background strain, C57Bl/6. We found the BPH/5 female mouse to be uniquely different from BPH/2 and C57Bl/6 mice with a genetically complex landscape. The majority of non-synonymous consequences within the coding region of BPH/5 females were missense mutations found most abundant on chromosome X when comparing BPH/5 and BPH/2, and on chromosome 8 when comparing BPH/5 to C57Bl/6. Genetic mutations in BPH/5 females largely belong to immune system-related processes, with overlap between BPH/5 and BPH/2 models. Further studies examining each gene mutation during pregnancy are warranted to determine key contributors to the BPH/5 preeclamptic-like phenotype and to identify genetic similarities to women that develop preeclampsia.
Collapse
|