1
|
Roessler J, Zimmermann F, Heidecker B, Landmesser U, Haghikia A. Gut microbiota-related modulation of immune mechanisms in post-infarction remodelling and heart failure. ESC Heart Fail 2024. [PMID: 39385474 DOI: 10.1002/ehf2.14991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 10/12/2024] Open
Abstract
The immune system has long been recognized as a key driver in the progression of heart failure (HF). However, clinical trials targeting immune effectors have consistently failed to improve patient outcome across different HF aetiologies. The activation of the immune system in HF is complex, involving a broad network of pro-inflammatory and immune-modulating components, which complicates the identification of specific immune pathways suitable for therapeutic targeting. Increasing attention has been devoted to identifying gut microbial pathways that affect cardiac remodelling and metabolism and, thereby impacting the development of HF. In particular, gut microbiota-derived metabolites, absorbed by the host and transported to the peripheral circulation, can act as signalling molecules, influencing metabolism and immune homeostasis. Recent reports suggest that the gut microbiota plays a crucial role in modulating immune processes involved in HF. Here, we summarize recent advances in understanding the contributory role of gut microbiota in (auto-)immune pathways that critically determine the progression or alleviation of HF. We also thoroughly discuss potential gut microbiota-based intervention strategies to treat or decelerate HF progression.
Collapse
Affiliation(s)
- Johann Roessler
- University Hospital St Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
| | - Friederike Zimmermann
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
| | - Bettina Heidecker
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin, Berlin Institute of Health (BIH), Berlin, Germany
| | - Arash Haghikia
- University Hospital St Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin, Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
2
|
Kalupahana NS, Moustaid-Moussa N. Beyond blood pressure, fluid and electrolyte homeostasis - Role of the renin angiotensin aldosterone system in the interplay between metabolic diseases and breast cancer. Acta Physiol (Oxf) 2024; 240:e14164. [PMID: 38770946 DOI: 10.1111/apha.14164] [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: 01/16/2024] [Revised: 04/16/2024] [Accepted: 04/29/2024] [Indexed: 05/22/2024]
Abstract
The classical renin angiotensin aldosterone system (RAAS), as well as the recently described counter-regulatory or non-canonical RAAS have been well characterized for their role in cardiovascular homeostasis. Moreover, extensive research has been conducted over the past decades on both paracrine and the endocrine roles of local RAAS in various metabolic regulations and in chronic diseases. Clinical evidence from patients on RAAS blockers as well as pre-clinical studies using rodent models of genetic manipulations of RAAS genes documented that this system may play important roles in the interplay between metabolic diseases and cancer, namely breast cancer. Some of these studies suggest potential therapeutic applications and repurposing of RAAS inhibitors for these diseases. In this review, we discuss the mechanisms by which RAAS is involved in the pathogenesis of metabolic diseases such as obesity and type-2 diabetes as well as the role of this system in the initiation, expansion and/or progression of breast cancer, especially in the context of metabolic diseases.
Collapse
Affiliation(s)
- Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
3
|
Deng F, Lei J, Chen J, Zhao M, Zhao C, Fu M, Sun M, Zhang M, Qiu J, Gao Q. DNA methylation-mediated 11βHSD2 downregulation drives the increases in angiotensin-converting enzyme and angiotensin II within preeclamptic placentas. FASEB J 2024; 38:e23714. [PMID: 38814727 DOI: 10.1096/fj.202400199r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/01/2024]
Abstract
Preeclampsia (PE) is a complex human-specific complication frequently associated with placental pathology. The local renin-angiotensin system (RAS) in the human placenta, which plays a crucial role in regulating placental function, has been extensively documented. Glucocorticoids (GCs) are a class of steroid hormones. PE cases often have abnormalities in GCs levels and placental GCs barrier. Despite extensive speculation, there is currently no robust evidence indicating that GCs regulate placental RAS. This study aims to investigate these potential relationships. Plasma and placental samples were collected from both normal and PE pregnancies. The levels of angiotensin-converting enzyme (ACE), angiotensin II (Ang II), cortisol, and 11β-hydroxysteroid dehydrogenases (11βHSD) were analyzed. In PE placentas, cortisol, ACE, and Ang II levels were elevated, while 11βHSD2 expression was reduced. Interestingly, a positive correlation was observed between ACE and cortisol levels in the placenta. A significant inverse correlation was found between the methylation statuses within the 11βHSD2 gene promoter and its expression, meanwhile, 11βHSD2 expression was negatively correlated with cortisol and ACE levels. In vitro experiments using placental trophoblast cells confirmed that active GCs can stimulate ACE transcription and expression through the GR pathway. Furthermore, 11βHSD2 knockdown could enhance this activating effect. An in vivo study using a rat model of intrauterine GCs overexposure during mid-to-late gestation suggested that excess GCs in utero lead to increased ACE and Ang II levels in the placenta. Collectively, this study provides the first evidence of the relationships between 11βHSD2 expression, GCs barrier, ACE, and Ang II levels in the placenta. It not only contributes to understanding the pathological features of the placental GCs barrier and RAS under PE conditions, also provides important information for revealing the pathological mechanism of PE.
Collapse
Affiliation(s)
- Fengying Deng
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Jiahui Lei
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Jie Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Meng Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
- Department of Obstetrics and Gynecology, the Third People's Hospital of Bengbu Affiliated to Bengbu Medical College, Bengbu, Anhui, China
| | - Chenxuan Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Mengyu Fu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Miao Sun
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Meihua Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Junlan Qiu
- Department of Oncology and Hematology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, P.R. China
| | - Qinqin Gao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| |
Collapse
|
4
|
Li CX, Yue L. The Multifaceted Nature of Macrophages in Cardiovascular Disease. Biomedicines 2024; 12:1317. [PMID: 38927523 PMCID: PMC11201197 DOI: 10.3390/biomedicines12061317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/01/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
As the leading cause of mortality worldwide, cardiovascular disease (CVD) represents a variety of heart diseases and vascular disorders, including atherosclerosis, aneurysm, ischemic injury in the heart and brain, arrythmias, and heart failure. Macrophages, a diverse population of immune cells that can promote or suppress inflammation, have been increasingly recognized as a key regulator in various processes in both healthy and disease states. In healthy conditions, these cells promote the proper clearance of cellular debris, dead and dying cells, and provide a strong innate immune barrier to foreign pathogens. However, macrophages can play a detrimental role in the progression of disease as well, particularly those inflammatory in nature. This review will focus on the current knowledge regarding the role of macrophages in cardiovascular diseases.
Collapse
Affiliation(s)
- Cindy X. Li
- Department of Cell Biology, Pat and Jim Calhoun Cardiovascular Center, University of Connecticut Health Center, Farmington, CT 06030, USA;
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Lixia Yue
- Department of Cell Biology, Pat and Jim Calhoun Cardiovascular Center, University of Connecticut Health Center, Farmington, CT 06030, USA;
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
5
|
De C, Xuan L, jingjing Z, Honghong Z, Kun Z, Song D, Yaqi S, Ying J, Cheng C, Jian L. Analysis of changes in high-mobility group box 1, receptor for advanced glycation endproducts, and T helper 17/regulatory T balance in severe preeclampsia with acute heart failure. J Clin Hypertens (Greenwich) 2024; 26:431-440. [PMID: 38523455 PMCID: PMC11007805 DOI: 10.1111/jch.14784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 03/26/2024]
Abstract
We measured the levels of High-Mobility Group Box 1 (HMGB1), Receptor for Advanced Glycation Endproducts (RAGE), T Helper 17 cells (Th17), Regulatory T cells (Treg), and related cytokines in the peripheral blood of patients with severe preeclampsia (SPE) complicated with acute heart failure (AHF) to explore the expression changes in these indicators. In total, 96 patients with SPE admitted to Gansu Provincial Maternity and Child-care Hospital between June 2020 and June 2022 were included in the study. The patients were divided into SPE+AHF (40 patients) and SPE (56 patients) groups based on whether they suffered from AHF. Additionally, 56 healthy pregnant women who either received prenatal examinations or were admitted to our hospital for delivery during the same period were selected as the healthy control group. An enzyme-linked immunosorbent assay was performed to detect the expression levels of HMGB1, RAGE, interleukin (IL)-17, IL-6, transforming growth factor β (TGF-β), IL-10, and NT-proBNP in plasma. Flow cytometry was employed to determine the percentages of Th17 and Treg cells. Compared to the healthy control group, the SPE+AHF and SPE groups had higher plasma levels of HMGB1 and RAGE expression, higher Th17 percentage and Th17/Treg ratio, and lower Treg percentage. Compared to the SPE group, the SPE+AHF group had higher plasma levels of HMGB1 and RAGE expression, higher Th17 percentage and Th17/Treg ratio, and lower Treg percentage (P < .05). In patients with SPE with AHF, plasma HMGB1 was positively correlated with RAGE, Th17, Th17/Treg, IL-17, and IL-6 and was negatively correlated with TGF-β and IL-10 (P < .05). Our findings revealed that patients with SPE with AHF had elevated levels of HMGB1 and RAGE while exhibiting Th17/Treg immune imbalance, suggesting that the abnormal expression of these indicators may be involved in the pathogenesis of SPE with AHF.
Collapse
Affiliation(s)
- Chen De
- First Clinical Medical SchoolLanzhou UniversityLanzhouChina
- Emergency Medical CenterGansu Provincial Maternity and Child‐care HospitalLanzhouChina
| | - Liang Xuan
- Department of AllergyGansu Provincial Maternity and Child‐care HospitalLanzhouChina
| | - Zhang jingjing
- Medical Genetics CenterGansu Provincial Maternity and Child‐care HospitalLanzhouChina
| | - Zhang Honghong
- Emergency Medical CenterGansu Provincial Maternity and Child‐care HospitalLanzhouChina
| | - Zuo Kun
- Emergency Medical CenterGansu Provincial Maternity and Child‐care HospitalLanzhouChina
| | - Du Song
- Emergency Medical CenterGansu Provincial Maternity and Child‐care HospitalLanzhouChina
| | - Song Yaqi
- Emergency Medical CenterGansu Provincial Maternity and Child‐care HospitalLanzhouChina
| | - Jiang Ying
- Emergency Medical CenterGansu Provincial Maternity and Child‐care HospitalLanzhouChina
| | - Cheng Cheng
- Emergency Medical CenterGansu Provincial Maternity and Child‐care HospitalLanzhouChina
| | - Liu Jian
- First Clinical Medical SchoolLanzhou UniversityLanzhouChina
- Emergency Medical CenterGansu Provincial Maternity and Child‐care HospitalLanzhouChina
| |
Collapse
|
6
|
Dai Y, Shen Z, Khachatryan LG, Vadiyan DE, Karampoor S, Mirzaei R. Unraveling mechanistic insights into the role of microbiome in neurogenic hypertension: A comprehensive review. Pathol Res Pract 2023; 249:154740. [PMID: 37567034 DOI: 10.1016/j.prp.2023.154740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023]
Abstract
Neurogenic hypertension, a complex and multifactorial cardiovascular disorder, is known to be influenced by various genetic, environmental, and lifestyle factors. In recent years, there has been growing interest in the role of the gut microbiome in hypertension pathogenesis. The bidirectional communication between the gut microbiota and the central nervous system, known as the microbiota-gut-brain axis, has emerged as a crucial mechanism through which the gut microbiota exerts its influence on neuroinflammation, immune responses, and blood pressure regulation. Recent studies have shown how the microbiome has a substantial impact on a variety of physiological functions, such as cardiovascular health. The increased sympathetic activity to the gut may cause microbial dysbiosis, increased permeability of the gut, and increased inflammatory reactions by altering a number of intestinal bacteria producing short-chain fatty acids (SCFAs) and the concentrations of lipopolysaccharide (LPS) in the plasma. Collectively, these microbial metabolic and structural compounds stimulate sympathetic stimulation, which may be an important stage in the onset of hypertension. The result is an upsurge in peripheral and central inflammatory response. In addition, it has recently been shown that a link between the immune system and the gut microbiota might play a significant role in hypertension. The therapeutic implications of the gut microbiome including probiotic usage, prebiotics, dietary modifications, and fecal microbiota transplantation in neurogenic hypertension have also been found. A large body of research suggests that probiotic supplementation might help reduce chronic inflammation and hypertension that have an association with dysbiosis in the gut microbiota. Overall, this review sheds light on the intricate interplay between the gut microbiome and neurogenic hypertension, providing valuable insights for both researchers and clinicians. As our knowledge of the microbiome's role in hypertension expands, novel therapeutic strategies and diagnostic biomarkers may pave the way for more effective management and prevention of this prevalent cardiovascular disorder. Exploring the potential of the microbiome in hypertension offers an exciting avenue for future research and offers opportunities for precision medicine and improved patient care.
Collapse
Affiliation(s)
- Yusang Dai
- Physical Examination Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Zheng Shen
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Lusine G Khachatryan
- Department of Pediatric Diseases, N.F. Filatov Clinical Institute of Children's Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Russia
| | - Diana E Vadiyan
- Institute of Dentistry, Department of Pediatric, Preventive Dentistry and Orthodontics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Russia
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
7
|
Evnouchidou I, Koumantou D, Nugue M, Saveanu L. M1-aminopeptidase family - beyond antigen-trimming activities. Curr Opin Immunol 2023; 83:102337. [PMID: 37216842 DOI: 10.1016/j.coi.2023.102337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 05/24/2023]
Abstract
Antigen (Ag)-trimming aminopeptidases belong to the oxytocinase subfamily of M1 metallopeptidases. In humans, this subfamily contains the endoplasmic reticulum aminopeptidases 1 and 2 (ERAP1 and 2) and the insulin-responsive aminopeptidase (IRAP, synonym oxytocinase), an endosomal enzyme. The ability of these enzymes to trim antigenic precursors and to generate major histocompatibility class-I ligands has been demonstrated extensively for ERAP1, less for ERAP2, which is absent in rodents, and exclusively in the context of cross-presentation for IRAP. During 20 years of research on these aminopeptidases, their enzymatic function has been very well characterized and their genetic association with autoimmune diseases, cancers, and infections is well established. The mechanisms by which these proteins are associated to human diseases are not always clear. This review discusses the Ag-trimming-independent functions of the oxytocinase subfamily of M1 aminopeptidases and the new questions raised by recent publications on IRAP and ERAP2.
Collapse
Affiliation(s)
- Irini Evnouchidou
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France; CNRS ERL8252, Paris, France; Université de Paris, Site Xavier Bichat, Paris, France; Inflamex Laboratory of Excellence, Paris, France; Inovarion, Paris, France
| | - Despoina Koumantou
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France; CNRS ERL8252, Paris, France; Université de Paris, Site Xavier Bichat, Paris, France; Inflamex Laboratory of Excellence, Paris, France
| | - Mathilde Nugue
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France; CNRS ERL8252, Paris, France; Université de Paris, Site Xavier Bichat, Paris, France; Inflamex Laboratory of Excellence, Paris, France
| | - Loredana Saveanu
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France; CNRS ERL8252, Paris, France; Université de Paris, Site Xavier Bichat, Paris, France; Inflamex Laboratory of Excellence, Paris, France.
| |
Collapse
|
8
|
da Silva MC, dos Santos VM, da Silva MVB, Prazeres TCMM, Cartágenes MDSS, Calzerra NTM, de Queiroz TM. Involvement of shedding induced by ADAM17 on the nitric oxide pathway in hypertension. Front Mol Biosci 2022; 9:1032177. [PMID: 36310604 PMCID: PMC9614329 DOI: 10.3389/fmolb.2022.1032177] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/04/2022] [Indexed: 11/15/2022] Open
Abstract
A Disintegrin and Metalloprotease 17 (ADAM17), also called tumor necrosis factor-ɑ (TNF-ɑ) convertase (TACE), is a well-known protease involved in the sheddase of growth factors, chemokines and cytokines. ADAM17 is also enrolled in hypertension, especially by shedding of angiotensin converting enzyme type 2 (ACE2) leading to impairment of angiotensin 1–7 [Ang-(1–7)] production and injury in vasodilation, induction of renal damage and cardiac hypertrophy. Activation of Mas receptor (MasR) by binding of Ang-(1–7) induces an increase in the nitric oxide (NO) gaseous molecule, which is an essential factor of vascular homeostasis and blood pressure control. On the other hand, TNF-ɑ has demonstrated to stimulate a decrease in nitric oxide bioavailability, triggering a disrupt in endothelium-dependent vasorelaxation. In spite of the previous studies, little knowledge is available about the involvement of the metalloprotease 17 and the NO pathways. Here we will provide an overview of the role of ADAM17 and Its mechanisms implicated with the NO formation.
Collapse
Affiliation(s)
- Mirelly Cunha da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| | - Vanessa Maria dos Santos
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| | - Matheus Vinícius B. da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| | | | | | | | - Thyago Moreira de Queiroz
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
- *Correspondence: Thyago Moreira de Queiroz,
| |
Collapse
|
9
|
Satou R, Franco M, Dugas CM, Katsurada A, Navar LG. Immunosuppression by Mycophenolate Mofetil Mitigates Intrarenal Angiotensinogen Augmentation in Angiotensin II-Dependent Hypertension. Int J Mol Sci 2022; 23:ijms23147680. [PMID: 35887028 PMCID: PMC9319385 DOI: 10.3390/ijms23147680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/01/2022] [Accepted: 07/07/2022] [Indexed: 02/01/2023] Open
Abstract
Augmentation of intrarenal angiotensinogen (AGT) leads to further formation of intrarenal angiotensin II (Ang II) and the development of hypertensive kidney injury. Recent studies demonstrated that macrophages and the enhanced production of pro-inflammatory cytokines can be crucial mediators of renal AGT augmentation in hypertension. Accordingly, this study investigated the effects of immunosuppression by mycophenolate mofetil (MMF) on intrarenal AGT augmentation. Ang II (80 ng/min) was infused with or without daily administration of MMF (50 mg/kg) to Sprague-Dawley rats for 2 weeks. Mean arterial pressure (MAP) in Ang II infused rats was slightly higher (169.7 ± 6.1 mmHg) than the Ang II + MMF group (154.7 ± 2.0 mmHg), but was not statistically different from the Ang II + MMF group. MMF treatment suppressed Ang II-induced renal macrophages and IL-6 elevation. Augmentation of urinary AGT by Ang II infusion was attenuated by MMF treatment (control: 89.3 ± 25.2, Ang II: 1194 ± 305.1, and Ang II + MMF: 389 ± 192.0 ng/day). The augmentation of urinary AGT by Ang II infusion was observed before the onset of proteinuria. Elevated intrarenal AGT mRNA and protein levels in Ang II infused rats were also normalized by the MMF treatment (AGT mRNA, Ang II: 2.5 ± 0.2 and Ang II + MMF: 1.5 ± 0.1, ratio to control). Ang II-induced proteinuria, mesangial expansion and renal tubulointerstitial fibrosis were attenuated by MMF. Furthermore, MMF treatment attenuated the augmentation of intrarenal NLRP3 mRNA, a component of inflammasome. These results indicate that stimulated cytokine production in macrophages contributes to intrarenal AGT augmentation in Ang II-dependent hypertension, which leads to the development of kidney injury.
Collapse
Affiliation(s)
- Ryousuke Satou
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA; (C.M.D.); (A.K.); (L.G.N.)
- Correspondence: ; Tel.: +1-504-988-4364
| | - Martha Franco
- Departments of Nephrology and Pathology, Instituto Nacional de Cardiologia, Mexico City 14080, Mexico;
| | - Courtney M. Dugas
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA; (C.M.D.); (A.K.); (L.G.N.)
| | - Akemi Katsurada
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA; (C.M.D.); (A.K.); (L.G.N.)
| | - L. Gabriel Navar
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA; (C.M.D.); (A.K.); (L.G.N.)
| |
Collapse
|
10
|
Savedchuk S, Raslan R, Nystrom S, Sparks MA. Emerging Viral Infections and the Potential Impact on Hypertension, Cardiovascular Disease, and Kidney Disease. Circ Res 2022; 130:1618-1641. [PMID: 35549373 DOI: 10.1161/circresaha.122.320873] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Viruses are ubiquitous in the environment and continue to have a profound impact on human health and disease. The COVID-19 pandemic has highlighted this with impressive morbidity and mortality affecting the world's population. Importantly, the link between viruses and hypertension, cardiovascular disease, and kidney disease has resulted in a renewed focus and attention on this potential relationship. The virus responsible for COVID-19, SARS-CoV-2, has a direct link to one of the major enzymatic regulatory systems connected to blood pressure control and hypertension pathogenesis, the renin-angiotensin system. This is because the entry point for SARS-CoV-2 is the ACE2 (angiotensin-converting enzyme 2) protein. ACE2 is one of the main enzymes responsible for dampening the primary effector peptide Ang II (angiotensin II), metabolizing it to Ang-(1-7). A myriad of clinical questions has since emerged and are covered in this review. Several other viruses have been linked to hypertension, cardiovascular disease, and kidney health. Importantly, patients with high-risk apolipoprotein L1 (APOL1) alleles are at risk for developing the kidney lesion of collapsing glomerulopathy after viral infection. This review will highlight several emerging viruses and their potential unique tropisms for the kidney and cardiovascular system. We focus on SARS-CoV-2 as this body of literature in regards to cardiovascular disease has advanced significantly since the COVID-19 pandemic.
Collapse
Affiliation(s)
- Solomiia Savedchuk
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC (S.S., S.N., M.A.S.)
| | - Rasha Raslan
- Internal Medicine, Virginia Commonwealth University, Richmond (R.R.)
| | - Sarah Nystrom
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC (S.S., S.N., M.A.S.)
| | - Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC (S.S., S.N., M.A.S.)
- Renal Section, Durham VA Health Care System, NC (M.A.S.)
| |
Collapse
|
11
|
Intestinal Microbiota-Derived Short Chain Fatty Acids in Host Health and Disease. Nutrients 2022; 14:nu14091977. [PMID: 35565943 PMCID: PMC9105144 DOI: 10.3390/nu14091977] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/01/2022] [Accepted: 05/06/2022] [Indexed: 01/27/2023] Open
Abstract
Intestinal microbiota has its role as an important component of human physiology. It produces metabolites that module key functions to establish a symbiotic crosstalk with their host. Among them, short chain fatty acids (SCFAs), produced by intestinal bacteria during the fermentation of partially and non-digestible polysaccharides, play key roles in regulating colon physiology and changing intestinal environment. Recent research has found that SCFAs not only influence the signal transduction pathway in the gut, but they also reach tissues and organs outside of the gut, through their circulation in the blood. Growing evidence highlights the importance of SCFAs level in influencing health maintenance and disease development. SCFAs are probably involved in the management of host health in a complicated (positive or negative) way. Here, we review the current understanding of SCFAs effects on host physiology and discuss the potential prevention and therapeutics of SCFAs in a variety of disorders. It provides a systematic theoretical basis for the study of mechanisms and precise intake level of SCFAs to promote human health.
Collapse
|
12
|
Luo M, Mou Q, Liu L, Tian J, Liu L. Treg/Th17 Ratio Regulation May Play an Important Role in Epigallocatechin-3-Gallate-Mediated Attenuation of Increased Afterload-Induced Cardiac Hypertrophy. J Cardiovasc Pharmacol 2022; 79:711-718. [PMID: 35058409 PMCID: PMC9067088 DOI: 10.1097/fjc.0000000000001220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/23/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT The aim of this study was to investigate whether Treg/Th17 ratio regulation plays an important role in epigallocatechin-3-gallate (EGCG) in attenuating increased afterload-induced cardiac hypertrophy. Three-month-old male C57BL/6 mice were divided into sham + vehicle, abdominal aortic constriction (AAC) + vehicle, and AAC + EGCG groups. Intraperitoneal EGCG (50 mg/kg/d) administration was conducted. Cardiac structure and function were examined by ultrasonography. Pathology was examined by hematoxylin and eosin staining, wheat germ agglutinin staining, and Masson's trichome staining. T-lymphocyte subtypes were analyzed using immunofluorescence and flow cytometry assays. Ultrasonography showed that the ventricular wall in the AAC + vehicle group was thicker than that in the sham + vehicle group (P < 0.05). Hematoxylin and eosin staining revealed cardiomyocyte hypertrophy accompanied by a small amount of inflammatory cell infiltration in the AAC + vehicle group. The results of wheat germ agglutinin staining demonstrated the presence of hypertrophic cardiomyocytes in the AAC + vehicle group (P < 0.01). Masson's trichome staining showed cardiac fibrosis in the AAC + vehicle group, and the immunofluorescence assay revealed infiltration of CD4+ cells in both AAC + vehicle and AAC + EGCG groups. Splenic flow cytometry showed a significant increase in the proportion of Treg cells in the AAC + EGCG group (P < 0.05). The proportion of Th17 cells in the AAC + vehicle group was significantly higher than that in the sham + vehicle group (P < 0.05). In conclusion, changes in the Treg/Th17 ratio are associated with the occurrence of myocardial hypertrophy caused by increased afterload. Moreover, regulation of the Treg/Th17 ratio by EGCG may play an important role in the attenuation of myocardial hypertrophy.
Collapse
Affiliation(s)
- Min Luo
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China; and
| | - Qiuhong Mou
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China; and
| | - Lingjuan Liu
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China; and
| | - Jie Tian
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China; and
| | - Lifei Liu
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China; and
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
13
|
Pan M, Vasbinder A, Anderson E, Catalan T, Shadid HR, Berlin H, Padalia K, O’Hayer P, Meloche C, Azam TU, Khaleel I, Michaud E, Blakely P, Bitar A, Huang Y, Zhao L, Pop‐Busui R, Loosen SH, Chalkias A, Tacke F, Giamarellos‐Bourboulis EJ, Reiser J, Eugen‐Olsen J, Hayek SS. Angiotensin-Converting Enzyme Inhibitors, Angiotensin II Receptor Blockers, and Outcomes in Patients Hospitalized for COVID-19. J Am Heart Assoc 2021; 10:e023535. [PMID: 34889102 PMCID: PMC9075226 DOI: 10.1161/jaha.121.023535] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/19/2021] [Indexed: 01/28/2023]
Abstract
Background Use of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers (ACEi/ARB) is thought to affect COVID-19 through modulating levels of angiotensin-converting enzyme 2, the cell entry receptor for SARS-CoV2. We sought to assess the association between ACEi/ARB, biomarkers of inflammation, and outcomes in patients hospitalized for COVID-19. Methods and Results We leveraged the ISIC (International Study of Inflammation in COVID-19), identified patients admitted for symptomatic COVID-19 between February 1, 2020 and June 1, 2021 for COVID-19, and examined the association between in-hospital ACEi/ARB use and all-cause death, need for ventilation, and need for dialysis. We estimated the causal effect of ACEi/ARB on the composite outcomes using marginal structural models accounting for serial blood pressure and serum creatinine measures. Of 2044 patients in ISIC, 1686 patients met inclusion criteria, of whom 398 (23.6%) patients who were previously on ACEi/ARB received at least 1 dose during their hospitalization for COVID-19. There were 215 deaths, 407 patients requiring mechanical ventilation, and 124 patients who required dialysis during their hospitalization. Prior ACEi/ARB use was associated with lower levels of soluble urokinase plasminogen activator receptor and C-reactive protein. In multivariable analysis, in-hospital ACEi/ARB use was associated with a lower risk of the composite outcome of in-hospital death, mechanical ventilation, or dialysis (adjusted hazard ratio 0.49, 95% CI [0.36-0.65]). Conclusions In patients hospitalized for COVID-19, ACEi/ARB use was associated with lower levels of inflammation and lower risk of in-hospital outcomes. Clinical trials will define the role of ACEi/ARB in the treatment of COVID-19. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT04818866.
Collapse
Affiliation(s)
- Michael Pan
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Alexi Vasbinder
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Elizabeth Anderson
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Toniemarie Catalan
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Husam R. Shadid
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Hanna Berlin
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Kishan Padalia
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Patrick O’Hayer
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Chelsea Meloche
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Tariq U. Azam
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Ibrahim Khaleel
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | | | - Pennelope Blakely
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Abbas Bitar
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Yiyuan Huang
- Department of BiostatisticsSchool of Public HealthUniversity of MichiganAnn ArborMI
| | - Lili Zhao
- Department of BiostatisticsSchool of Public HealthUniversity of MichiganAnn ArborMI
| | - Rodica Pop‐Busui
- Division of Metabolism, Endocrinology and DiabetesDepartment of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Sven H. Loosen
- Clinic for Gastroenterology, Hepatology and Infectious DiseasesMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - Athanasios Chalkias
- Department of AnesthesiologySchool of Health SciencesFaculty of MedicineUniversity of ThessalyLarisaGreece
- Outcomes Research ConsortiumClevelandOH
| | - Frank Tacke
- Department of Hepatology & GastroenterologyCampus Charité Mitte/Campus Virchow‐KlinikumCharité University Medicine BerlinBerlinGermany
| | | | - Jochen Reiser
- Department of MedicineRush University Medical CenterChicagoIL
| | - Jesper Eugen‐Olsen
- Department of Clinical ResearchCopenhagen University Hospital Amager and HvidovreHvidovreDenmark
| | - Salim S. Hayek
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMI
| | | |
Collapse
|
14
|
Gersh FL, O'Keefe JH, Lavie CJ, Henry BM. The Renin-Angiotensin-Aldosterone System in Postmenopausal Women: The Promise of Hormone Therapy. Mayo Clin Proc 2021; 96:3130-3141. [PMID: 34736778 DOI: 10.1016/j.mayocp.2021.08.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 07/29/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022]
Abstract
Estradiol (E2) plays an underrecognized role in modulating body-wide systems, including important interactions with the renin-angiotensin-aldosterone system (RAAS). The RAAS is an immunomodulating system that is critical for maintaining homeostasis across multiple organ systems. The diverse interactions between E2 and the RAAS help maintain cardiometabolic homeostasis, including successful physiologic responses to trauma and infectious pathogens. Estradiol deficiency (ie, menopause) results in impaired responses and increased susceptibility to infectious pathogens. Both immune and cardiometabolic function decline with reduced E2 production, in part because the RAAS becomes dysregulated by E2 deficiency, leaving RAAS predominantly in its proinflammatory state and predisposing to systemic low-grade inflammation. Estradiol deficiency and RAAS dysregulation contribute to impaired immune responses and increased incidence of cardiac hypertrophy, hypertension, atherosclerotic cardiovascular disease, arrhythmias, and heart failure. The RAAS consists of dual, counterbalancing pathways-proinflammatory and anti-inflammatory. Estradiol is a signaling agent that plays a major role in determining which RAAS pathway predominates. The proinflammatory pathway is activated early in response to infection or trauma, followed by up-regulation of the anti-inflammatory pathway, to resolve inflammation and to restore homeostasis. Estradiol influences activation of the "switch" to restore the anti-inflammatory state. The dysregulated RAAS is a primary target of current cardiovascular therapeutics focused on blocking portions of its proinflammatory pathway. However, RAAS-modifying pharmaceuticals often provide imperfect solutions to these physiologic disruptions and underscore the need for improved approaches to menopausal medicine. Estradiol therapy and optimal lifestyle practices combined with RAAS-modifying pharmaceuticals may be an ideal strategy to optimize postmenopausal health.
Collapse
Affiliation(s)
- Felice L Gersh
- University of Arizona School of Medicine, Division of Integrative Medicine, Tucson, LA
| | - James H O'Keefe
- Saint Luke's Mid America Heart Institute, University of Missouri-Kansas City, Kansas City, LA
| | - Carl J Lavie
- Ochsner Heart and Vascular Institute, Ochsner Clinical School-The University of Queensland School of Medicine, New Orleans, LA
| | - Brandon M Henry
- Cincinnati Children's Hospital, The Heart Institute, CICU, Cincinnati, OH.
| |
Collapse
|
15
|
Jia P, Huang B, You Y, Su H, Gao L. Ketogenic diet aggravates kidney dysfunction by exacerbating metabolic disorders and inhibiting autophagy in spontaneously hypertensive rats. Biochem Biophys Res Commun 2021; 573:13-18. [PMID: 34375764 DOI: 10.1016/j.bbrc.2021.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/02/2021] [Indexed: 11/26/2022]
Abstract
AIMS To assess the effects of a ketogenic diet on metabolism and renal fibrosis in spontaneously hypertensive rats. MATERIALS AND METHODS Male spontaneously hypertensive rats (SHRs) and Wistar Kyoto (WKY) rats were randomly divided into a ketogenic diet group and a normal diet group. Blood glucose and metabolites were measured after 4 weeks. Renal autophagy-related protein expression was detected by Western blot, and renal fibrosis was detected by Masson staining. RESULTS Compared with the normal diet, the ketogenic diet led to significantly decreased glucose tolerance and metabolism; overactivated the renin-angiotensin-aldosterone system; and reduced renal autophagy-related protein expression in SHRs; Masson staining and other experiments showed that the ketogenic diet had no significant effect on hypertensive renal fibrosis. CONCLUSION A Ketogenic diet could lead to disorders of glucose and lipid metabolism, increase hypertension by activating the RAAS, reduce renal autophagy levels and aggravate renal parenchymal damage. Therefore, a ketogenic diet, as a kind of natural therapy, should be vigilantly monitored to prevent further damage in patients with hypertension.
Collapse
Affiliation(s)
- Ping Jia
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Bi Huang
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yuehua You
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Su
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lingyun Gao
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
16
|
Pan K, Jiang S, Du X, Zeng X, Zhang J, Song L, Lei L, Zhou J, Kan H, Sun Q, Xie Y, Dong C, Zhao J. Parental PM 2 .5 exposure changes Th17/Treg cells in offspring, is associated with the elevation of blood pressure. ENVIRONMENTAL TOXICOLOGY 2021; 36:1152-1161. [PMID: 33605513 DOI: 10.1002/tox.23114] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/10/2020] [Accepted: 02/04/2021] [Indexed: 06/12/2023]
Abstract
Epidemiological evidences have indicated that fine particulate matter (PM2.5 ) exposure is associated with the occurrence and development of hypertension. The present study aims to explore the effects of parental PM2.5 exposure on blood pressure in offspring and elucidate the potential mechanism. The parental male and female C57BL/6 mice were exposed to concentrated PM2.5 or filtered air (FA) using Shanghai Meteorological and Environmental Animal Exposure System (Shanghai-METAS) for 16 weeks. At week 12, the mice were assigned to breed offspring. The male offspring mice were further exposed to PM2.5 or FA as above method. During the parental exposure, the average PM2.5 concentration was 133.7 ± 53.32 μg/m3 in PM chamber, whereas the average concentration in FA chamber was 9.4 ± 0.23 μg/m3 . Similarly, during the offspring exposure, the average concentration in PM and FA chamber were 100.76 ± 26.97 μg/m3 and 9.15 ± 0.15 μg/m3 , respectively. The PM2.5 -exposed offspring mice displayed the elevation of blood pressure, the increase of angiotensin II (Ang II), the decrease of angiotensin converting enzyme 2 (ACE2) and Ang (1-7) in serum when compared with the FA-exposed offspring mice. The similar results displayed in the proteins expression of ACE2, AT1R, and Ang (1-7) in vessel and kidney. More importantly, parental PM exposure further induced the increase in serous Ang II and the protein expression of AT1R in vessel, but decrease in ACE2 and Ang (1-7). The serous Ang II was positively associated with splenic T helper type 17 (Th17) cell population and serous IL (interleukin)-17A, but negatively associated with T regular (Treg) cell population and serous IL-10. The results suggested that parental air pollution exposure might induce the elevation of offspring blood pressure via mediate Th17- and Treg-related immune microenvironment.
Collapse
Affiliation(s)
- Kun Pan
- Department of Infectious Disease Control, Center for Disease Control and Prevention of Shangcheng in Hangzhou, Hangzhou, Zhejiang, China
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Shuo Jiang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Xihao Du
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Xuejiao Zeng
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Jia Zhang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Liying Song
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Lei Lei
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Ji Zhou
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai, China
| | - Haidong Kan
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Qinghua Sun
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, Ohio
| | - Yuquan Xie
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chen Dong
- Administrative office, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Jinzhuo Zhao
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai, China
| |
Collapse
|
17
|
Ying W, Tang K, Avolio E, Schilling JM, Pasqua T, Liu MA, Cheng H, Gao H, Zhang J, Mahata S, Ko MS, Bandyopadhyay G, Das S, Roth DM, Sahoo D, Webster NJG, Sheikh F, Ghosh G, Patel HH, Ghosh P, van den Bogaart G, Mahata SK. Immunosuppression of Macrophages Underlies the Cardioprotective Effects of CST (Catestatin). Hypertension 2021; 77:1670-1682. [PMID: 33826401 DOI: 10.1161/hypertensionaha.120.16809] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Wei Ying
- Department of Medicine (W.Y., E.A., M.A.L., H.G., J.Z., S.M., G.B., F.S., N.J.G.W., P.G., S.K.M.), University of California San Diego, La Jolla
| | - Kechun Tang
- VA San Diego Healthcare System, CA (T.P., K.T., J.M.S., D.M.R., N.J.G.W., H.H.P., S.K.M.)
| | - Ennio Avolio
- Department of Medicine (W.Y., E.A., M.A.L., H.G., J.Z., S.M., G.B., F.S., N.J.G.W., P.G., S.K.M.), University of California San Diego, La Jolla.,Comparative Anatomy & Cytology, Dept. of Biology, Ecology and Earth Science, University of Calabria, Arcavacata di Rende-Cosenza, Italy (E.A.)
| | - Jan M Schilling
- VA San Diego Healthcare System, CA (T.P., K.T., J.M.S., D.M.R., N.J.G.W., H.H.P., S.K.M.).,Department of Anesthesiology (J.M.S., D.M.R., H.H.P.), University of California San Diego, La Jolla
| | - Teresa Pasqua
- VA San Diego Healthcare System, CA (T.P., K.T., J.M.S., D.M.R., N.J.G.W., H.H.P., S.K.M.).,Department of Health Science, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy (T.P.)
| | - Matthew A Liu
- Department of Medicine (W.Y., E.A., M.A.L., H.G., J.Z., S.M., G.B., F.S., N.J.G.W., P.G., S.K.M.), University of California San Diego, La Jolla
| | - Hongqiang Cheng
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China (H.C.)
| | - Hong Gao
- Department of Medicine (W.Y., E.A., M.A.L., H.G., J.Z., S.M., G.B., F.S., N.J.G.W., P.G., S.K.M.), University of California San Diego, La Jolla
| | - Jing Zhang
- Department of Medicine (W.Y., E.A., M.A.L., H.G., J.Z., S.M., G.B., F.S., N.J.G.W., P.G., S.K.M.), University of California San Diego, La Jolla
| | - Sumana Mahata
- Department of Medicine (W.Y., E.A., M.A.L., H.G., J.Z., S.M., G.B., F.S., N.J.G.W., P.G., S.K.M.), University of California San Diego, La Jolla
| | - Myung S Ko
- Department of Chemistry and Biochemistry (M.S.K., G.G.), University of California San Diego, La Jolla
| | - Gautam Bandyopadhyay
- Department of Medicine (W.Y., E.A., M.A.L., H.G., J.Z., S.M., G.B., F.S., N.J.G.W., P.G., S.K.M.), University of California San Diego, La Jolla
| | - Soumita Das
- Department of Pathology (S.D.), University of California San Diego, La Jolla
| | - David M Roth
- VA San Diego Healthcare System, CA (T.P., K.T., J.M.S., D.M.R., N.J.G.W., H.H.P., S.K.M.).,Department of Anesthesiology (J.M.S., D.M.R., H.H.P.), University of California San Diego, La Jolla
| | - Debashis Sahoo
- Department of Pediatrics (D.S.), University of California San Diego, La Jolla.,Department of Computer Science and Engineering (D.S.), University of California San Diego, La Jolla
| | - Nicholas J G Webster
- VA San Diego Healthcare System, CA (T.P., K.T., J.M.S., D.M.R., N.J.G.W., H.H.P., S.K.M.).,Department of Medicine (W.Y., E.A., M.A.L., H.G., J.Z., S.M., G.B., F.S., N.J.G.W., P.G., S.K.M.), University of California San Diego, La Jolla
| | - Farah Sheikh
- Department of Medicine (W.Y., E.A., M.A.L., H.G., J.Z., S.M., G.B., F.S., N.J.G.W., P.G., S.K.M.), University of California San Diego, La Jolla
| | - Gourisankar Ghosh
- Department of Chemistry and Biochemistry (M.S.K., G.G.), University of California San Diego, La Jolla
| | - Hemal H Patel
- VA San Diego Healthcare System, CA (T.P., K.T., J.M.S., D.M.R., N.J.G.W., H.H.P., S.K.M.).,Department of Anesthesiology (J.M.S., D.M.R., H.H.P.), University of California San Diego, La Jolla
| | - Pradipta Ghosh
- Department of Medicine (W.Y., E.A., M.A.L., H.G., J.Z., S.M., G.B., F.S., N.J.G.W., P.G., S.K.M.), University of California San Diego, La Jolla.,Cellular and Molecular Medicine (P.G.), University of California San Diego, La Jolla
| | - Geert van den Bogaart
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, the Netherlands (G.v.d.B.).,Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands (G.v.d.B.)
| | - Sushil K Mahata
- VA San Diego Healthcare System, CA (T.P., K.T., J.M.S., D.M.R., N.J.G.W., H.H.P., S.K.M.).,Department of Medicine (W.Y., E.A., M.A.L., H.G., J.Z., S.M., G.B., F.S., N.J.G.W., P.G., S.K.M.), University of California San Diego, La Jolla
| |
Collapse
|
18
|
Basile DP, Ullah MM, Collet JA, Mehrotra P. T helper 17 cells in the pathophysiology of acute and chronic kidney disease. Kidney Res Clin Pract 2021; 40:12-28. [PMID: 33789382 PMCID: PMC8041630 DOI: 10.23876/j.krcp.20.185] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Both acute and chronic kidney disease have a strong underlying inflammatory component. This review focuses primarily on T helper 17 (Th17) cells as mediators of inflammation and their potential to modulate acute and chronic kidney disease. We provide updated information on factors and signaling pathways that promote Th17 cell differentiation with specific reference to kidney disease. We highlight numerous clinical studies that have investigated Th17 cells in the setting of human kidney disease and provide updated summaries from various experimental animal models of kidney disease indicating an important role for Th17 cells in renal fibrosis and hypertension. We focus on the pleiotropic effects of Th17 cells in different renal cell types as potentially relevant to the pathogenesis of kidney disease. Finally, we highlight studies that present contrasting roles for Th17 cells in kidney disease progression.
Collapse
Affiliation(s)
- David P Basile
- Department of Anatomy, Cell Biology & Physiology, Indiana University of Medicine, Indianapolis, IN, United States
| | - Md Mahbub Ullah
- Department of Anatomy, Cell Biology & Physiology, Indiana University of Medicine, Indianapolis, IN, United States
| | - Jason A Collet
- Department of Anatomy, Cell Biology & Physiology, Indiana University of Medicine, Indianapolis, IN, United States
| | - Purvi Mehrotra
- Department of Anatomy, Cell Biology & Physiology, Indiana University of Medicine, Indianapolis, IN, United States
| |
Collapse
|
19
|
Basile DP, Abais-Battad JM, Mattson DL. Contribution of Th17 cells to tissue injury in hypertension. Curr Opin Nephrol Hypertens 2021; 30:151-158. [PMID: 33394732 PMCID: PMC8221512 DOI: 10.1097/mnh.0000000000000680] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Hypertension has been demonstrated to be a chief contributor to morbidity and mortality throughout the world. Although the cause of hypertension is multifactorial, emerging evidence, obtained in experimental studies, as well as observational studies in humans, points to the role of inflammation and immunity. Many aspects of immune function have now been implicated in hypertension and end-organ injury; this review will focus upon the recently-described role of Th17 cells in this pathophysiological response. RECENT FINDINGS Studies in animal models and human genetic studies point to a role in the adaptive immune system as playing a contributory role in hypertension and renal tissue damage. Th17 cells, which produce the cytokine IL17, are strongly pro-inflammatory cells, which may contribute to tissue damage if expressed in chronic disease conditions. The activity of these cells may be enhanced by physiological factors associated with hypertension such as dietary salt or Ang II. This activity may culminate in the increased sodium retaining activity and exacerbation of inflammation and renal fibrosis via multiple cellular mechanisms. SUMMARY Th17 cells are a distinct component of the adaptive immune system that may strongly enhance pathways leading to increased sodium reabsorption, elevated vascular tone and end-organ damage. Moreover, this pathway may lend itself towards specific targeting for treatment of kidney disease and hypertension.
Collapse
Affiliation(s)
- David P Basile
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - David L Mattson
- Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA
| |
Collapse
|
20
|
Nistala R, Meuth AI, Smith C, An J, Habibi J, Hayden MR, Johnson M, Aroor A, Whaley-Connell A, Sowers JR, McKarns SC, Bender SB. DPP4 inhibition mitigates ANG II-mediated kidney immune activation and injury in male mice. Am J Physiol Renal Physiol 2021; 320:F505-F517. [PMID: 33522410 DOI: 10.1152/ajprenal.00565.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Recent evidence suggests that dipeptidyl peptidase-4 (DPP4) inhibition with saxagliptin (Saxa) is renoprotective under comorbid conditions associated with activation of the renin-angiotensin-aldosterone system (RAAS), such as diabetes, obesity, and hypertension, which confer a high cardiovascular risk. Immune system activation is now recognized as a contributor to RAAS-mediated tissue injury, and, importantly, immunomodulatory effects of DPP4 have been reported. Accordingly, we examined the hypothesis that DPP4 inhibition with Saxa attenuates angiotensin II (ANG II)-induced kidney injury and albuminuria via attenuation of immune activation in the kidney. To this end, male mice were infused with either vehicle or ANG II (1,000 ng/kg/min, s.c.) for 3 wk and received either placebo or Saxa (10 mg/kg/day, p.o.) during the final 2 wk. ANG II infusion increased kidney, but not plasma, DPP4 activity in vivo as well as DPP4 activity in cultured proximal tubule cells. The latter was prevented by angiotensin receptor blockade with olmesartan. Further, ANG II induced hypertension and kidney injury characterized by mesangial expansion, mitochondrial damage, reduced brush border megalin expression, and albuminuria. Saxa inhibited DPP4 activity ∼50% in vivo and attenuated ANG II-mediated kidney injury, independent of blood pressure. Further mechanistic experiments revealed mitigation by Saxa of proinflammatory and profibrotic mediators activated by ANG II in the kidney, including CD8+ T cells, resident macrophages (CD11bhiF4/80loLy6C-), and neutrophils. In addition, Saxa improved ANG II suppressed anti-inflammatory regulatory T cell and T helper 2 lymphocyte activity. Taken together, these results demonstrate, for the first time, blood pressure-independent involvement of renal DPP4 activation contributing to RAAS-dependent kidney injury and immune activation.NEW & NOTEWORTHY This work highlights the role of dipeptidyl peptidase-4 (DPP4) in promoting ANG II-mediated kidney inflammation and injury. Specifically, ANG II infusion in mice led to increases in blood pressure and kidney DPP4 activity, which then led to activation of CD8+ T cells, Ly6C- macrophages, and neutrophils and suppression of anti-inflammatory T helper 2 lymphocytes and regulatory T cells. Collectively, this led to kidney injury, characterized by mesangial expansion, mitochondrial damage, and albuminuria, which were mitigated by DPP4 inhibition independent of blood pressure reduction.
Collapse
Affiliation(s)
- Ravi Nistala
- Divisions of Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, Missouri.,Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Alex I Meuth
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Cassandra Smith
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Jianzhong An
- Divisions of Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, Missouri.,Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Javad Habibi
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - M R Hayden
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Megan Johnson
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Annayya Aroor
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Adam Whaley-Connell
- Divisions of Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, Missouri.,Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - James R Sowers
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Susan C McKarns
- Departments of Microbiology and Immunology and Surgery, University of Missouri School of Medicine, Columbia, Missouri
| | - Shawn B Bender
- Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
21
|
Ji X, Tan W, Zhang C, Zhai Y, Hsueh Y, Zhang Z, Zhang C, Lu Y, Duan B, Tan G, Na R, Deng G, Niu G. TWIRLS, a knowledge-mining technology, suggests a possible mechanism for the pathological changes in the human host after coronavirus infection via ACE2. Drug Dev Res 2020; 81:1004-1018. [PMID: 32657473 PMCID: PMC7404951 DOI: 10.1002/ddr.21717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/05/2020] [Accepted: 06/27/2020] [Indexed: 12/12/2022]
Abstract
Faced with the current large-scale public health emergency, collecting, sorting, and analyzing biomedical information related to the "SARS-CoV-2" should be done as quickly as possible to gain a global perspective, which is a basic requirement for strengthening epidemic control capacity. However, for human researchers studying viruses and hosts, the vast amount of information available cannot be processed effectively and in a timely manner, particularly if our scientific understanding is also limited, which further lowers the information processing efficiency. We present TWIRLS (Topic-wise inference engine of massive biomedical literatures), a method that can deal with various scientific problems, such as liver cancer, acute myeloid leukemia, and so forth, which can automatically acquire, organize, and classify information. Additionally, this information can be combined with independent functional data sources to build an inference system via a machine-based approach, which can provide relevant knowledge to help human researchers quickly establish subject cognition and to make more effective decisions. Using TWIRLS, we automatically analyzed more than three million words in more than 14,000 literature articles in only 4 hr. We found that an important regulatory factor angiotensin-converting enzyme 2 (ACE2) may be involved in host pathological changes on binding to the coronavirus after infection. On triggering functional changes in ACE2/AT2R, the cytokine homeostasis regulation axis becomes imbalanced via the Renin-Angiotensin System and IP-10, leading to a cytokine storm. Through a preliminary analysis of blood indices of COVID-19 patients with a history of hypertension, we found that non-ARB (Angiotensin II receptor blockers) users had more symptoms of severe illness than ARB users. This suggests ARBs could potentially be used to treat acute lung injury caused by coronavirus infection.
Collapse
Affiliation(s)
- Xiaoyang Ji
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Inner Mongolia Autonomous Region College of Animal ScienceInner Mongolia Agricultural UniversityHohhotChina
- Joint Turing‐Darwin Laboratory of Phil Rivers Technology Ltd. and Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- Department of Computational Biology, Phil Rivers Technology LtdBeijingChina
| | - Wenting Tan
- Department of Infectious DiseasesSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Chunming Zhang
- Joint Turing‐Darwin Laboratory of Phil Rivers Technology Ltd. and Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- Department of Computational Biology, Phil Rivers Technology LtdBeijingChina
- Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- West Institute of Computing TechnologyChinese Academy of SciencesChongqingChina
| | - Yubo Zhai
- Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yiching Hsueh
- Joint Turing‐Darwin Laboratory of Phil Rivers Technology Ltd. and Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- Department of Computational Biology, Phil Rivers Technology LtdBeijingChina
| | - Zhonghai Zhang
- Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
| | - Chunli Zhang
- Department of Computational Biology, Phil Rivers Technology LtdBeijingChina
| | - Yanqiu Lu
- Department of Infectious DiseasesChongqing Public Health Medical CenterChongqingChina
| | - Bo Duan
- Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- West Institute of Computing TechnologyChinese Academy of SciencesChongqingChina
| | - Guangming Tan
- Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- West Institute of Computing TechnologyChinese Academy of SciencesChongqingChina
| | - Renhua Na
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Inner Mongolia Autonomous Region College of Animal ScienceInner Mongolia Agricultural UniversityHohhotChina
| | - Guohong Deng
- Department of Infectious DiseasesSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Gang Niu
- Joint Turing‐Darwin Laboratory of Phil Rivers Technology Ltd. and Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- Department of Computational Biology, Phil Rivers Technology LtdBeijingChina
- West Institute of Computing TechnologyChinese Academy of SciencesChongqingChina
| |
Collapse
|
22
|
McCoy K, Peterson A, Tian Y, Sang Y. Immunogenetic Association Underlying Severe COVID-19. Vaccines (Basel) 2020; 8:E700. [PMID: 33233531 PMCID: PMC7711778 DOI: 10.3390/vaccines8040700] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/15/2020] [Accepted: 11/17/2020] [Indexed: 12/16/2022] Open
Abstract
SARS-CoV2 has caused the current pandemic of new coronavirus disease 2019 (COVID-19) worldwide. Clinical outcomes of COVID-19 illness range broadly from asymptotic and mild to a life-threatening situation. This casts uncertainties for defining host determinants underlying the disease severity. Recent genetic analyses based on extensive clinical sample cohorts using genome-wide association studies (GWAS) and high throughput sequencing curation revealed genetic errors and gene loci associated with about 20% of life-threatening COVID-19 cases. Significantly, most of these critical genetic loci are enriched in two immune signaling pathways, i.e., interferon-mediated antiviral signaling and chemokine-mediated/inflammatory signaling. In line with these genetic profiling studies, the broad spectrum of COVID-19 illness could be explained by immuno-pathological regulation of these critical immunogenetic pathways through various epigenetic mechanisms, which further interconnect to other vital components such as those in the renin-angiotensin-aldosterone system (RAAS) because of its direct interaction with the virus causing COVID-19. Together, key genes unraveled by genetic profiling may provide targets for precisely early risk diagnosis and prophylactic design to relieve severe COVID-19. The confounding epigenetic mechanisms may be key to understanding the clinical broadness of COVID-19 illness.
Collapse
Affiliation(s)
- Kendall McCoy
- Department of Biology, College of Life and Physical Sciences, Tennessee State University, 3500 John A. Merritt Boulevard, Nashville, TN 37209, USA; (K.M.); (A.P.)
| | - Autumn Peterson
- Department of Biology, College of Life and Physical Sciences, Tennessee State University, 3500 John A. Merritt Boulevard, Nashville, TN 37209, USA; (K.M.); (A.P.)
| | - Yun Tian
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, 3500 John A. Merritt Boulevard, Nashville, TN 37209, USA;
| | - Yongming Sang
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, 3500 John A. Merritt Boulevard, Nashville, TN 37209, USA;
| |
Collapse
|
23
|
Annweiler C, Cao Z, Wu Y, Faucon E, Mouhat S, Kovacic H, Sabatier JM. Counter-regulatory 'Renin-Angiotensin' System-based Candidate Drugs to Treat COVID-19 Diseases in SARS-CoV-2-infected Patients. Infect Disord Drug Targets 2020; 20:407-408. [PMID: 32418532 DOI: 10.2174/1871526520666200518073329] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Cédric Annweiler
- Department of Geriatric Medicine and Memory Clinic, Research Center on Autonomy and Longevity, University Hospital, Angers, France
| | - Zhijian Cao
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yingliang Wu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | | | | | - Hervé Kovacic
- Universite Aix-Marseille, Institut de Neuro-physiopathologie (INP), UMR 7051, Faculte de Pharmacie, 27 Bd Jean Moulin, 13385 Marseille Cedex, France
| | - Jean-Marc Sabatier
- Universite Aix-Marseille, Institut de Neuro-physiopathologie (INP), UMR 7051, Faculte de Pharmacie, 27 Bd Jean Moulin, 13385 Marseille Cedex, France
| |
Collapse
|
24
|
Stein RA, Young LM. From ACE2 to COVID-19: A multiorgan endothelial disease. Int J Infect Dis 2020; 100:425-430. [PMID: 32896660 PMCID: PMC7832810 DOI: 10.1016/j.ijid.2020.08.083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Affiliation(s)
- Richard A Stein
- NYU Tandon School of Engineering, Department of Chemical and Biomolecular Engineering, 6 MetroTech Center, Brooklyn, NY 11201, USA; LaGuardia Community College, Department of Natural Sciences, City University of New York, New York, NY 11101, USA.
| | - Lauren M Young
- University of Chicago, Department of Internal Medicine, 5841 S Maryland Ave, Chicago, IL 60637, USA.
| |
Collapse
|
25
|
Bansal R, Gubbi S, Muniyappa R. Metabolic Syndrome and COVID 19: Endocrine-Immune-Vascular Interactions Shapes Clinical Course. Endocrinology 2020; 161:bqaa112. [PMID: 32603424 PMCID: PMC7337756 DOI: 10.1210/endocr/bqaa112] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic is caused by the novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Individuals with metabolic syndrome are at increased risk for poor disease outcomes and mortality from COVID-19. The pathophysiologic mechanisms for these observations have not been fully elucidated. A critical interaction between SARS-CoV-2 and the angiotensin-converting enzyme 2 (ACE2) facilitates viral entry into the host cell. ACE2 is expressed in pancreatic islets, vascular endothelium, and adipose tissue, and the SARS-CoV-2 -ACE2 interaction in these tissues, along with other factors, governs the spectrum and the severity of clinical manifestations among COVID-19 patients with metabolic syndrome. Moreover, the pro-inflammatory milieu observed in patients with metabolic syndrome may contribute toward COVID-19-mediated host immune dysregulation, including suboptimal immune responses, hyperinflammation, microvascular dysfunction, and thrombosis. This review describes the spectrum of clinical features, the likely pathophysiologic mechanisms, and potential implications for the management of metabolic syndrome in COVID-19 patients.
Collapse
Affiliation(s)
- Rashika Bansal
- Clinical Endocrine Section, Diabetes, Endocrinology, and
Obesity Branch, National Institute of Diabetes and Digestive and Kidney
Diseases, National Institutes of Health, Bethesda, MD
| | - Sriram Gubbi
- Clinical Endocrine Section, Diabetes, Endocrinology, and
Obesity Branch, National Institute of Diabetes and Digestive and Kidney
Diseases, National Institutes of Health, Bethesda, MD
| | - Ranganath Muniyappa
- Clinical Endocrine Section, Diabetes, Endocrinology, and
Obesity Branch, National Institute of Diabetes and Digestive and Kidney
Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
26
|
Guo F, Qiu X, Zhu Y, Tan Z, Li Z, Ouyang D. Circulating choline is associated with coronary artery stenosis in patients with hypertension: A cross-sectional study of Chinese adults. J Clin Hypertens (Greenwich) 2020; 22:2069-2076. [PMID: 32966687 DOI: 10.1111/jch.14025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 01/08/2023]
Abstract
Choline is an important nutrient involved in multiple biosynthesis pathways. However, whether circulating choline levels are associated with the risk of hypertension (HTN) and artery stenosis in HTN remains unknown. We investigated the correlations of plasma choline with HTN and coronary artery injury and explored the utility of plasma choline as a diagnostic biomarker for HTN and artery stenosis. 193 HTN patients and 154 age- and sex-matched healthy controls (CON) were recruited in this study. Fasting plasma choline was detected using liquid chromatography tandem mass spectrometry. Choline levels were significantly higher in HTN without artery stenosis (HTN-AS) than CON (8.07 [7.19-9.24] μM vs 7.03 [6.21-8.13] μM, P < .01) group and were further upregulated in HTN with artery stenosis (HTN + AS) (8.63 [7.09-10.59] μM, P < .01) group. Patients with multivessel disease (MVD) also exhibited higher choline levels than those with single vessel disease (SVD) (8.64 [7.16-10.55] μM vs 8.04(6.74-9.38) μM, P < .01). Increased choline levels were independently associated with the risk of HTN (OR = 1.2, 95% CI: 1-1.45, P = .05), HTN + AS (OR = 1.27, 95% CI: 1.09-1.48, P < .01), and MVD (OR = 1.16, 95% CI: 1.02-1.31, P = .02) after adjustment for multiple risk factors. Receiver operating characteristic (ROC) analysis showed that choline had an area under curve (AUC) score of 0.69, 0.67, and 0.63 in determining HTN, HTN + AS, and MVD. In conclusion, higher choline levels were associated with increased risk of HTN and artery stenosis in hypertensive patients.
Collapse
Affiliation(s)
- Fei Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China.,Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, China
| | - Xueting Qiu
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanting Zhu
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zhirong Tan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Zhenyu Li
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Dongsheng Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China.,Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, China
| |
Collapse
|
27
|
Stone E, Kiat H, McLachlan CS. Atrial fibrillation in COVID-19: A review of possible mechanisms. FASEB J 2020; 34:11347-11354. [PMID: 33078484 DOI: 10.1096/fj.202001613] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022]
Abstract
A relationship between COVID-19 infection and an increasing incidence of atrial fibrillation has been observed. However, the underlying pathophysiology as a precipitant to AF has not been reviewed. This paper will consider the possible pathological and immunological AF mechanisms as a result, of COVID-19 infection. We discuss the role myocardial microvascular pericytes expressing the ACE-2 receptor and their potential for an organ-specific cardiac involvement with COVID-19. Dysfunctional microvascular support by pericytes or endothelial cells may increase the propensity for AF via increased myocardial inflammation, fibrosis, increased tissue edema, and interstitial hydrostatic pressure. All of these factors can lead to electrical perturbances at the tissue and cellular level. We also consider the contribution of Angiotensin, pulmonary hypertension, and regulatory T cells as additional contributors to AF during COVID-19 infection. Finally, reference is given to two common drugs, corticosteroids and metformin, in COVID-19 and how they might influence AF incidence.
Collapse
Affiliation(s)
- Elijah Stone
- Health Vertical, Centre for Healthy Futures, Torrens University Australia, Sydney, NSW, Australia
| | - Hosen Kiat
- Health Vertical, Centre for Healthy Futures, Torrens University Australia, Sydney, NSW, Australia.,Cardiac Health Institute, Eastwood, NSW, Australia.,The Australian School of Advanced Medicine, 2 Technology Place, Macquarie University, Sydney, NSW, Australia
| | - Craig S McLachlan
- Health Vertical, Centre for Healthy Futures, Torrens University Australia, Sydney, NSW, Australia
| |
Collapse
|
28
|
de Queiroz TM, Lakkappa N, Lazartigues E. ADAM17-Mediated Shedding of Inflammatory Cytokines in Hypertension. Front Pharmacol 2020; 11:1154. [PMID: 32848763 PMCID: PMC7403492 DOI: 10.3389/fphar.2020.01154] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022] Open
Abstract
The increase of Angiontesin-II (Ang-II), one of the key peptides of the renin-angiotensin system (RAS), and its binding to the Ang-II type 1 receptor (AT1R) during hypertension is a crucial mechanism leading to AD\AM17 activation. Among the reported membrane anchored proteins cleaved by ADAM17, immunological cytokines (TNF-α, IFN-γ, TGF-β, IL-4, IL-10, IL-13, IL-6, FKN) are the major class of substrates, modulation of which triggers inflammation. The rise in ADAM17 levels has both central and peripheral implications in inflammation-mediated hypertension. This narrative review provides an overview of the role of ADAM17, with a special focus on its cellular regulation on neuronal and peripheral inflammation-mediated hypertension. Finally, it highlights the importance of ADAM17 with regards to the biology of inflammatory cytokines and their roles in hypertension.
Collapse
Affiliation(s)
- Thyago M. de Queiroz
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco - UFPE, Vitória de Santo Antão, Brazil
| | - Navya Lakkappa
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, United States
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, United States
| |
Collapse
|
29
|
Uso terapéutico de los inhibidores de la enzima convertidora de angiotensina en pacientes con COVID-19: las «dos caras de la moneda». REVISTA COLOMBIANA DE CARDIOLOGÍA 2020. [PMCID: PMC7365126 DOI: 10.1016/j.rccar.2020.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
La evidencia actual es limitada para determinar el impacto del uso de los inhibidores de la enzima convertidora de angiotensina (IECA) en la predisposición al empeoramiento de la enfermedad del coronavirus 2019 (COVID-19). Inicialmente se reportó que en los pacientes con progresión grave de la COVID-19 existía una mortalidad elevada, los cuales tenían antecedentes de hipertensión arterial, diabetes mellitus, enfermedad cardiovascular y enfermedad renal crónica. Parte de estos pacientes también tenía en común que utilizaban IECA, lo cual alertó a la comunidad médica sobre su riesgo potencial en coexistencia con COVID-19. Sin embargo, estudios más recientes de casos-controles encontraron que los inhibidores del sistema renina-angiotensina, incluyendo los IECA, no incrementan el riesgo de COVID-19 o de requerir admisión hospitalaria por esta causa. Diferentes revistas científicas han facilitado el acceso a reportes preliminares, dejando a discreción de la comunidad médica y científica hacer uso de dicha información para promover el desarrollo de estudios que confirmen experimentalmente dichos hallazgos, preclínicos y epidemiológicos, que finalmente impacten en las decisiones de la práctica clínica para beneficiar a los pacientes con COVID-19. En esta revisión de la literatura se exploran los diferentes efectos mediados por los IECA que podrían estar relacionados con la respuesta inmune durante la infección y la transmisión de COVID-19, compilando evidencia disponible que evalúa si en realidad representan un riesgo o si, por el contrario, confieren un efecto protector.
Collapse
|
30
|
MiR155-5p Inhibits Cell Migration and Oxidative Stress in Vascular Smooth Muscle Cells of Spontaneously Hypertensive Rats. Antioxidants (Basel) 2020; 9:antiox9030204. [PMID: 32121598 PMCID: PMC7140008 DOI: 10.3390/antiox9030204] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/23/2020] [Accepted: 02/26/2020] [Indexed: 02/06/2023] Open
Abstract
Migration of vascular smooth muscle cells (VSMCs) is essential for vascular reconstruction in hypertension and several vascular diseases. Our recent study showed that extracellular vesicles derived from vascular adventitial fibroblasts of normal rats inhibited VSMC proliferation by delivering miR155-5p to VSMCs. It is unknown whether miR155-5p inhibits cell migration and oxidative stress in VSMCs of spontaneously hypertensive rats (SHR) and in angiotensin II (Ang II)-treated VSMCs. The purpose of this study was to determine the role of miR155-5p in VSMC migration and its underlying mechanisms. Primary VSMCs were isolated from the aortic media of Wistar-Kyoto rats (WKY) and SHR. Wound healing assay and Boyden chamber assay were used to evaluate VSMC migration. A miR155-5p mimic inhibited, and a miR155-5p inhibitor promoted the migration of VSMC of SHR but had no significant effect on the migration of VSMC of WKY. The miR155-5p mimic inhibited angiotensin-converting enzyme (ACE) mRNA and protein expression in VSMCs. It also reduced superoxide anion production, NAD(P)H oxidase (NOX) activity, as well as NOX2, interleukin-1β (IL-1β), and tumor necrosis factor α (TNF-α) expression levels in VSMCs of SHR but not in VSMCs of WKY rats. Overexpression of miR155-5p inhibited VSMC migration and superoxide anion and IL-1β production in VSMCs of SHR but had no impact on exogenous Ang II-induced VSMC migration and on superoxide anion and IL-1β production in WKY rats and SHR. These results indicate that miR155-5p inhibits VSMC migration in SHR by suppressing ACE expression and its downstream production of Ang II, superoxide anion, and inflammatory factors. However, miR155-5p had no effects on exogenous Ang II-induced VSMC migration.
Collapse
|
31
|
Wen Y, Rudemiller NP, Zhang J, Lu X, Ren J, Privratsky JR, Griffiths R, Zhang JJ, Hammer GE, Crowley SD. C-C Motif Chemokine Receptor 7 Exacerbates Hypertension Through Effects on T Lymphocyte Trafficking. Hypertension 2020; 75:869-876. [PMID: 31983306 DOI: 10.1161/hypertensionaha.119.14148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Activated T lymphocytes that infiltrate blood pressure control organs make a critical contribution to the pathogenesis of hypertension. Dendritic cells act as potent antigen-presenting cells to stimulate prohypertensive T cells. However, the mechanisms that facilitate the recruitment of prohypertensive T cells and dendritic cells into the kidney's draining lymph node during hypertension require elucidation. As CCR7 (C-C motif chemokine receptor type 7) directs the homing of lymphocytes and dendritic cells into lymph nodes, we posited that dendritic cell-mediated T lymphocyte stimulation in the renal lymph node is CCR7 dependent and required for a full hypertensive response. We found that CCR7-deficient (CCR7 KO) mice had a blunted hypertensive response in our model of chronic Ang II (angiotensin II) infusion. Ang II-infused CCR7 KO animals had exaggerated accumulation of CD8+ T cells in the kidney but reduced numbers of CD4+ and CD8+ T cells in the kidney's draining lymph node. To understand whether CCR7-dependent homing of T lymphocytes or dendritic cells into the lymph node regulates the hypertensive response, we injected CCR7 KO or wild-type T cells or dendritic cells into CCR7 KO recipients, neither of which restored the full hypertensive response to Ang II infusion. However, adoptive transfer of wild-type but not CCR7 KO T lymphocytes into RAG1 (recombination-activating gene 1)-deficient mice that lack a lymphocyte niche restored full blood pressure elevation during Ang II infusion. Thus, CCR7-dependent interactions between T lymphocytes and dendritic cells are essential for T lymphocyte stimulation and hypertension accruing from inappropriate activation of the renin-angiotensin system.
Collapse
Affiliation(s)
- Yi Wen
- From the Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Center, NC (Y.W., N.P.R., J.Z., X.L., J.R., R.G., S.D.C.)
| | - Nathan P Rudemiller
- From the Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Center, NC (Y.W., N.P.R., J.Z., X.L., J.R., R.G., S.D.C.)
| | - Jiandong Zhang
- From the Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Center, NC (Y.W., N.P.R., J.Z., X.L., J.R., R.G., S.D.C.)
| | - Xiaohan Lu
- From the Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Center, NC (Y.W., N.P.R., J.Z., X.L., J.R., R.G., S.D.C.)
| | - Jiafa Ren
- From the Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Center, NC (Y.W., N.P.R., J.Z., X.L., J.R., R.G., S.D.C.)
| | - Jamie R Privratsky
- Departments of Anesthesiology (J.R.P.), Duke University Medical Center, Durham, NC
| | - Robert Griffiths
- From the Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Center, NC (Y.W., N.P.R., J.Z., X.L., J.R., R.G., S.D.C.)
| | - Junyi J Zhang
- Immunology (J.J.Z., G.H.), Duke University Medical Center, Durham, NC
| | - Gianna E Hammer
- Immunology (J.J.Z., G.H.), Duke University Medical Center, Durham, NC.,Molecular Genetics and Microbiology (G.H.), Duke University Medical Center, Durham, NC
| | - Steven D Crowley
- From the Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Center, NC (Y.W., N.P.R., J.Z., X.L., J.R., R.G., S.D.C.)
| |
Collapse
|
32
|
Bartolomaeus H, Balogh A, Yakoub M, Homann S, Markó L, Höges S, Tsvetkov D, Krannich A, Wundersitz S, Avery EG, Haase N, Kräker K, Hering L, Maase M, Kusche-Vihrog K, Grandoch M, Fielitz J, Kempa S, Gollasch M, Zhumadilov Z, Kozhakhmetov S, Kushugulova A, Eckardt KU, Dechend R, Rump LC, Forslund SK, Müller DN, Stegbauer J, Wilck N. Short-Chain Fatty Acid Propionate Protects From Hypertensive Cardiovascular Damage. Circulation 2019; 139:1407-1421. [PMID: 30586752 PMCID: PMC6416008 DOI: 10.1161/circulationaha.118.036652] [Citation(s) in RCA: 443] [Impact Index Per Article: 88.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Arterial hypertension and its organ sequelae show characteristics of T cell–mediated inflammatory diseases. Experimental anti-inflammatory therapies have been shown to ameliorate hypertensive end-organ damage. Recently, the CANTOS study (Canakinumab Antiinflammatory Thrombosis Outcome Study) targeting interleukin-1β demonstrated that anti-inflammatory therapy reduces cardiovascular risk. The gut microbiome plays a pivotal role in immune homeostasis and cardiovascular health. Short-chain fatty acids (SCFAs) are produced from dietary fiber by gut bacteria and affect host immune homeostasis. Here, we investigated effects of the SCFA propionate in 2 different mouse models of hypertensive cardiovascular damage. Methods: To investigate the effect of SCFAs on hypertensive cardiac damage and atherosclerosis, wild-type NMRI or apolipoprotein E knockout–deficient mice received propionate (200 mmol/L) or control in the drinking water. To induce hypertension, wild-type NMRI mice were infused with angiotensin II (1.44 mg·kg–1·d–1 subcutaneous) for 14 days. To accelerate the development of atherosclerosis, apolipoprotein E knockout mice were infused with angiotensin II (0.72 mg·kg–1·d–1 subcutaneous) for 28 days. Cardiac damage and atherosclerosis were assessed using histology, echocardiography, in vivo electrophysiology, immunofluorescence, and flow cytometry. Blood pressure was measured by radiotelemetry. Regulatory T cell depletion using PC61 antibody was used to examine the mode of action of propionate. Results: Propionate significantly attenuated cardiac hypertrophy, fibrosis, vascular dysfunction, and hypertension in both models. Susceptibility to cardiac ventricular arrhythmias was significantly reduced in propionate-treated angiotensin II–infused wild-type NMRI mice. Aortic atherosclerotic lesion area was significantly decreased in propionate-treated apolipoprotein E knockout–deficient mice. Systemic inflammation was mitigated by propionate treatment, quantified as a reduction in splenic effector memory T cell frequencies and splenic T helper 17 cells in both models, and a decrease in local cardiac immune cell infiltration in wild-type NMRI mice. Cardioprotective effects of propionate were abrogated in regulatory T cell–depleted angiotensin II–infused mice, suggesting the effect is regulatory T cell–dependent. Conclusions: Our data emphasize an immune-modulatory role of SCFAs and their importance for cardiovascular health. The data suggest that lifestyle modifications leading to augmented SCFA production could be a beneficial nonpharmacological preventive strategy for patients with hypertensive cardiovascular disease.
Collapse
Affiliation(s)
- Hendrik Bartolomaeus
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., J.B., R.D., S.K.F., D.N.M., N.W.).,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., A.B., L.M., S.W., E.G.A., N.H., K.K., J.F., R.D., D.N.M., N.W.).,Berlin Institute of Health, Germany (H.B., A.B., L.M., E.G.A., N.H., K.K., R.D., S.K.F., D.N.M., N.W.)
| | - András Balogh
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., J.B., R.D., S.K.F., D.N.M., N.W.).,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., A.B., L.M., S.W., E.G.A., N.H., K.K., J.F., R.D., D.N.M., N.W.).,Berlin Institute of Health, Germany (H.B., A.B., L.M., E.G.A., N.H., K.K., R.D., S.K.F., D.N.M., N.W.)
| | - Mina Yakoub
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Germany (M.Y., S. Höges, L.H., L.C.R., J.S.)
| | - Susanne Homann
- Institute of Pharmacology and Clinical Pharmacology, University Hospital, Universitätsrat, Düsseldorf, Germany (S. Homann, M.G.)
| | - Lajos Markó
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., J.B., R.D., S.K.F., D.N.M., N.W.).,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., A.B., L.M., S.W., E.G.A., N.H., K.K., J.F., R.D., D.N.M., N.W.).,Berlin Institute of Health, Germany (H.B., A.B., L.M., E.G.A., N.H., K.K., R.D., S.K.F., D.N.M., N.W.)
| | - Sascha Höges
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Germany (M.Y., S. Höges, L.H., L.C.R., J.S.)
| | - Dmitry Tsvetkov
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., J.B., R.D., S.K.F., D.N.M., N.W.).,Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics and Interfaculty Center of Pharmacogenomics and Drug Research, Tübingen, Germany (D.T.)
| | - Alexander Krannich
- Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.)
| | - Sebastian Wundersitz
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., J.B., R.D., S.K.F., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., A.B., L.M., S.W., E.G.A., N.H., K.K., J.F., R.D., D.N.M., N.W.)
| | - Ellen G Avery
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., J.B., R.D., S.K.F., D.N.M., N.W.).,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., A.B., L.M., S.W., E.G.A., N.H., K.K., J.F., R.D., D.N.M., N.W.).,Berlin Institute of Health, Germany (H.B., A.B., L.M., E.G.A., N.H., K.K., R.D., S.K.F., D.N.M., N.W.)
| | - Nadine Haase
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., A.B., L.M., S.W., E.G.A., N.H., K.K., J.F., R.D., D.N.M., N.W.).,Berlin Institute of Health, Germany (H.B., A.B., L.M., E.G.A., N.H., K.K., R.D., S.K.F., D.N.M., N.W.)
| | - Kristin Kräker
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., A.B., L.M., S.W., E.G.A., N.H., K.K., J.F., R.D., D.N.M., N.W.).,Berlin Institute of Health, Germany (H.B., A.B., L.M., E.G.A., N.H., K.K., R.D., S.K.F., D.N.M., N.W.)
| | - Lydia Hering
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Germany (M.Y., S. Höges, L.H., L.C.R., J.S.)
| | - Martina Maase
- Institute of Physiology II, University of Münster, Germany (M.M., K.K.-V.)
| | | | - Maria Grandoch
- Institute of Pharmacology and Clinical Pharmacology, University Hospital, Universitätsrat, Düsseldorf, Germany (S. Homann, M.G.)
| | - Jens Fielitz
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., J.B., R.D., S.K.F., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., A.B., L.M., S.W., E.G.A., N.H., K.K., J.F., R.D., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Greifswald (J.F.)
| | - Stefan Kempa
- Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.).,Integrative Proteomics and Metabolomics Platform, Berlin Institute for Medical Systems Biology, Germany (S. Kempa)
| | - Maik Gollasch
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Medizinische Klinik mit Schwerpunkt Nephrologie und Internistische Intensivmedizin Charité - Universitätsmedizin Berlin, Germany (M.G., K.-U.E., N.W.)
| | - Zhaxybay Zhumadilov
- National Laboratory Astana Nazarbayev University, Kazakhstan (Z.Z., S. Kozhakhmetov, A. Kushugalova)
| | - Samat Kozhakhmetov
- National Laboratory Astana Nazarbayev University, Kazakhstan (Z.Z., S. Kozhakhmetov, A. Kushugalova)
| | - Almagul Kushugulova
- National Laboratory Astana Nazarbayev University, Kazakhstan (Z.Z., S. Kozhakhmetov, A. Kushugalova)
| | - Kai-Uwe Eckardt
- Medizinische Klinik mit Schwerpunkt Nephrologie und Internistische Intensivmedizin Charité - Universitätsmedizin Berlin, Germany (M.G., K.-U.E., N.W.)
| | - Ralf Dechend
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., J.B., R.D., S.K.F., D.N.M., N.W.).,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., A.B., L.M., S.W., E.G.A., N.H., K.K., J.F., R.D., D.N.M., N.W.).,Berlin Institute of Health, Germany (H.B., A.B., L.M., E.G.A., N.H., K.K., R.D., S.K.F., D.N.M., N.W.).,Department of Cardiology and Nephrology, HELIOS-Klinikum, Berlin, Germany (R.D.)
| | - Lars Christian Rump
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Germany (M.Y., S. Höges, L.H., L.C.R., J.S.)
| | - Sofia K Forslund
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., J.B., R.D., S.K.F., D.N.M., N.W.).,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.).,Berlin Institute of Health, Germany (H.B., A.B., L.M., E.G.A., N.H., K.K., R.D., S.K.F., D.N.M., N.W.).,European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany (S.K.F.)
| | - Dominik N Müller
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., J.B., R.D., S.K.F., D.N.M., N.W.).,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., A.B., L.M., S.W., E.G.A., N.H., K.K., J.F., R.D., D.N.M., N.W.).,Berlin Institute of Health, Germany (H.B., A.B., L.M., E.G.A., N.H., K.K., R.D., S.K.F., D.N.M., N.W.)
| | - Johannes Stegbauer
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Germany (M.Y., S. Höges, L.H., L.C.R., J.S.)
| | - Nicola Wilck
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., N.H., K.K., J.F., M.G., R.D., S.K.F., D.N.M., N.W.).,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., A.B., L.M., D.T., S.W., E.G.A., J.B., R.D., S.K.F., D.N.M., N.W.).,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (H.B., A.B., L.M., A. Krannich, E.G.A., N.H., K.K., S. Kempa, R.D., S.K.F., D.N.M., N.W.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., A.B., L.M., S.W., E.G.A., N.H., K.K., J.F., R.D., D.N.M., N.W.).,Berlin Institute of Health, Germany (H.B., A.B., L.M., E.G.A., N.H., K.K., R.D., S.K.F., D.N.M., N.W.).,Medizinische Klinik mit Schwerpunkt Nephrologie und Internistische Intensivmedizin Charité - Universitätsmedizin Berlin, Germany (M.G., K.-U.E., N.W.)
| |
Collapse
|
33
|
Role of complement 3 in the pathogenesis of hypertension. Hypertens Res 2019; 43:255-262. [DOI: 10.1038/s41440-019-0371-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/08/2019] [Accepted: 11/10/2019] [Indexed: 02/06/2023]
|
34
|
Carnagarin R, Matthews V, Zaldivia MTK, Peter K, Schlaich MP. The bidirectional interaction between the sympathetic nervous system and immune mechanisms in the pathogenesis of hypertension. Br J Pharmacol 2019; 176:1839-1852. [PMID: 30129037 PMCID: PMC6534787 DOI: 10.1111/bph.14481] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/26/2018] [Accepted: 08/05/2018] [Indexed: 12/14/2022] Open
Abstract
Over the last few years, evidence has accumulated to suggest that hypertension is, at least in part, an immune-mediated inflammatory disorder. Many links between immunity and hypertension have been established and provide a complex framework of mechanistic interactions contributing to the rise in BP. These include immune-mediated inflammatory processes affecting regulatory brain nuclei and interactions with other mediators of cardiovascular regulation such as the sympathetic nervous system. Sympathoexcitation differentially regulates T-cells based upon activation status of the immune cell as well as the resident organ. Exogenous and endogenous triggers activate signalling pathways in innate and adaptive immune cells resulting in pro-inflammatory cytokine production and activation of T-lymphocytes in the cardiovascular and renal regions, now considered major factors in the development of essential hypertension. The inflammatory cascade is sustained and exacerbated by the immune flow via the brain-bone marrow-spleen-gastrointestinal axis and thereby further aggravating immune-mediated pathways resulting in a vicious cycle of established hypertension and target organ damage. This review summarizes the evidence and recent advances in linking immune-mediated inflammation, sympathetic activation and their bidirectional interactions with the development of hypertension. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
- Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital UnitThe University of Western AustraliaPerthWAAustralia
| | - Vance Matthews
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital UnitThe University of Western AustraliaPerthWAAustralia
| | - Maria T K Zaldivia
- Atherothrombosis and Vascular BiologyBaker Heart and Diabetes InstituteMelbourneVicAustralia
- Department of MedicineMonash University, Royal Perth HospitalPerthWAAustralia
| | - Karlheinz Peter
- Atherothrombosis and Vascular BiologyBaker Heart and Diabetes InstituteMelbourneVicAustralia
- Department of MedicineMonash University, Royal Perth HospitalPerthWAAustralia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital UnitThe University of Western AustraliaPerthWAAustralia
- Department of CardiologyRoyal Perth HospitalPerthWAAustralia
- Department of NephrologyRoyal Perth HospitalPerthWAAustralia
| |
Collapse
|
35
|
Ganguly A, Sharma K, Majumder K. Food-derived bioactive peptides and their role in ameliorating hypertension and associated cardiovascular diseases. ADVANCES IN FOOD AND NUTRITION RESEARCH 2019; 89:165-207. [PMID: 31351525 DOI: 10.1016/bs.afnr.2019.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Non-communicable diseases including cardiovascular diseases (CVDs) and associated metabolic disorders are responsible for nearly 40 million deaths globally per year. Hypertension or high blood pressure (BP) is one of the primary reasons for the development of CVDs. A healthy nutritional strategy complementing with physical activity can substantially reduce high BP and prevent the occurrence of CVD-associated morbidity and mortality. Bioactive peptides currently are the next wave of the promising bench to clinic options for potential targeting chronic and acute health issues including hypertension. Peptides demonstrating anti-inflammatory, anti-oxidant, and angiotensin-converting enzyme-I inhibitory activity are widely studied for the amelioration of hypertension and associated CVDs. Isolating these potent bioactive peptides from different food sources is a promising endeavor toward nutraceutical based dietary management and prevention of hypertension. Understanding the pathophysiology of hypertension and the action mechanisms of the bioactive peptides would complement in designing and characterizing more potent peptides and suitable comprehensive dietary plans for the prevention of hypertension and associated CVDs.
Collapse
Affiliation(s)
- Advaita Ganguly
- Comprehensive Tissue Centre, UAH Transplant Services, Alberta Health Services, Edmonton, AB, Canada
| | - Kumakshi Sharma
- Health, Safety and Environment Branch, National Research Council Canada, Edmonton, AB, Canada
| | - Kaustav Majumder
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, United States.
| |
Collapse
|
36
|
Liu K, Liu C, Zhang Z. lncRNA GAS5 acts as a ceRNA for miR‐21 in suppressing PDGF‐bb‐induced proliferation and migration in vascular smooth muscle cells. J Cell Biochem 2019; 120:15233-15240. [DOI: 10.1002/jcb.28789] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Kefeng Liu
- School of Basic Medical Sciences Xiangnan University Chenzhou P.R. China
| | - Chen Liu
- School of Basic Medical Sciences Xiangnan University Chenzhou P.R. China
| | - Zhengyu Zhang
- Guangzhou Institute of Cardiovascular Disease The Second Affiliated Hospital of Guangzhou Medical University Guangzhou P.R. China
| |
Collapse
|
37
|
Tong Y, Ye C, Ren XS, Qiu Y, Zang YH, Xiong XQ, Wang JJ, Chen Q, Li YH, Kang YM, Zhu GQ. Exosome-Mediated Transfer of ACE (Angiotensin-Converting Enzyme) From Adventitial Fibroblasts of Spontaneously Hypertensive Rats Promotes Vascular Smooth Muscle Cell Migration. Hypertension 2019; 72:881-888. [PMID: 30354715 DOI: 10.1161/hypertensionaha.118.11375] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Migration of vascular smooth muscle cells (VSMCs) is pivotal for vascular remodeling in hypertension. Vascular adventitial fibroblasts (AFs) are important in the homeostasis of vascular structure. This study is designed to investigate the roles of AF exosomes (AFE) in VSMC migration and underling mechanism. Primary VSMCs and AFs were obtained from the aorta of spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats. VSMC migration was evaluated with Boyden chamber assay and wound healing assay. AFE from WKY rats and SHR were isolated and identified. AFE from SHR promoted but AFE from WKY rats had no significant effect on VSMC migration. The effects of AFE on VSMC migration were prevented by an exosome inhibitor GW4869, an AT1R (Ang II [angiotensin II] type 1 receptor) antagonist losartan, or an inhibitor of ACE (angiotensin-converting enzyme) captopril. ACE contents and activity were much higher in AFE from SHR than those from WKY rats. There were no significant difference in Ang II and AT1R mRNA and protein levels between AFE from SHR and AFE from WKY rats. AFE from SHR increased Ang II and ACE contents and ACE activity in VSMCs of WKY rats and SHR. The changes of Ang II contents and ACE activity were prevented by captopril. ACE knockdown in AFs reduced ACE contents and activity in AFE from SHR and inhibited AFE-induced migration of VSMCs of WKY rats and those of SHR. These results indicate that exosomes from AFs of SHR transfer ACE to VSMCs, which increases Ang II levels and activates AT1R in VSMCs and thereby promotes VSMC migration.
Collapse
Affiliation(s)
- Ying Tong
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | - Chao Ye
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | - Xing-Sheng Ren
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | - Yun Qiu
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | - Ying-Hao Zang
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | - Xiao-Qing Xiong
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | | | - Qi Chen
- Department of Pathophysiology (Q.C., Y.-H.L., G.-Q.Z.), Nanjing Medical University, Jiangsu, China
| | - Yue-Hua Li
- Department of Pathophysiology (Q.C., Y.-H.L., G.-Q.Z.), Nanjing Medical University, Jiangsu, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Cardiovascular Research Center, Xi'an Jiaotong University School of Medicine, China (Y.-M.K.)
| | - Guo-Qing Zhu
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.).,Department of Pathophysiology (Q.C., Y.-H.L., G.-Q.Z.), Nanjing Medical University, Jiangsu, China
| |
Collapse
|
38
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 663] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
39
|
Satou R, Penrose H, Navar LG. Inflammation as a Regulator of the Renin-Angiotensin System and Blood Pressure. Curr Hypertens Rep 2018; 20:100. [PMID: 30291560 DOI: 10.1007/s11906-018-0900-0] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW Mechanisms facilitating progression of hypertension via cross stimulation of the renin-angiotensin system (RAS) and inflammation have been proposed. Accordingly, we review and update evidence for regulation of RAS components by pro-inflammatory factors. RECENT FINDINGS Angiotensin II (Ang II), which is produced by RAS, induces vasoconstriction and consequent blood pressure elevation. In addition to this direct action, chronically elevated Ang II stimulates several pathophysiological mechanisms including generation of oxidative stress, stimulation of the nervous system, alterations in renal hemodynamics, and activation of the immune system. In particular, an activated immune system has been shown to contribute to the development of hypertension. Recent studies have demonstrated that immune cell-derived pro-inflammatory cytokines regulate RAS components, further accelerating systemic and local Ang II formation. Specifically, regulation of angiotensinogen (AGT) production by pro-inflammatory cytokines in the liver and kidney is proposed as a key mechanism underlying the progression of Ang II-dependent hypertension.
Collapse
Affiliation(s)
- Ryousuke Satou
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, 1430 Tulane Avenue, SL39, New Orleans, LA, 70112-2699, USA.
| | - Harrison Penrose
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, 1430 Tulane Avenue, SL39, New Orleans, LA, 70112-2699, USA
| | - L Gabriel Navar
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, 1430 Tulane Avenue, SL39, New Orleans, LA, 70112-2699, USA
| |
Collapse
|
40
|
Small HY, Migliarino S, Czesnikiewicz-Guzik M, Guzik TJ. Hypertension: Focus on autoimmunity and oxidative stress. Free Radic Biol Med 2018; 125:104-115. [PMID: 29857140 DOI: 10.1016/j.freeradbiomed.2018.05.085] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 12/25/2022]
Abstract
Understanding the causal role of the immune and inflammatory responses in hypertension has led to questions regarding the links between hypertension and autoimmunity. Immune pathology in primary hypertension mimics several autoimmune mechanisms observed in the pathogenesis of systemic lupus erythematosus, psoriasis, systemic sclerosis, rheumatoid arthritis and periodontitis. More importantly, the prevalence of hypertension in patients with these autoimmune diseases is significantly increased, when compared to control populations. Clinical and epidemiological evidence is reviewed along with possible mechanisms linking hypertension and autoimmunity. Inflammation and oxidative stress are linked in a self-perpetuating cycle that significantly contributes to the vascular dysfunction and renal damage associated with hypertension. T cell, B cell, macrophage and NK cell infiltration into these organs is essential for this pathology. Effector cytokines such as IFN-γ, TNF-α and IL-17 affect Na+/H+ exchangers in the kidney. In blood vessels, they lead to endothelial dysfunction and loss of nitric oxide bioavailability and cause vasoconstriction. Both renal and vascular effects are, in part, mediated through induction of reactive oxygen species-producing enzymes such as superoxide anion generating NADPH oxidases and dysfunction of anti-oxidant systems. These mechanisms have recently become important therapeutic targets of novel therapies focused on scavenging oxidative (isolevuglandin) modification of neo-antigenic peptides. Effects of classical immune targeted therapies focused on immunosuppression and anti-cytokine treatments are also reviewed.
Collapse
Affiliation(s)
- Heather Y Small
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Serena Migliarino
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Marta Czesnikiewicz-Guzik
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK; Department of Dental Prophylaxis and Experimental Dentistry, Dental School of Jagiellonian University, Krakow, Poland
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Department of Internal and Agricultural Medicine, Jagiellonian University Collegium Medicum, Krakow, Poland.
| |
Collapse
|
41
|
Cardiovascular Interactions between Fibroblast Growth Factor-23 and Angiotensin II. Sci Rep 2018; 8:12398. [PMID: 30120363 PMCID: PMC6098163 DOI: 10.1038/s41598-018-30098-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 07/14/2018] [Indexed: 12/12/2022] Open
Abstract
Both the activation of the renin angiotensin aldosterone system (RAAS) and elevations of circulating Fibroblast Growth Factor-23 (FGF-23) have been implicated in the pathogenesis of left ventricular hypertrophy (LVH) in chronic kidney disease. To investigate potential cross-talk between RAAS and FGF-23, we administered angiotensin II (Ang II) to wild-type rodents and the Hyp mouse model of excess FGF-23. Ang II administration for four weeks to wild-type rodents resulted in significant increases in systolic blood pressure and LVH. Unexpectedly, FGF-23 circulating levels were increased by 1.5-1.7 fold in Ang II treated animals. In addition, Ang II treatment increased expression of FGF-23 message levels in bone, the predominant tissue for FGF-23 production, and induced expression of FGF-23 and its co-receptor α-Klotho in the heart, which normally does not express FGF-23 or α-Klotho in physiologically relevant levels. Hyp mice with elevated FGF-23 exhibited increased blood pressure and LVH at baseline. Ang II administration to Hyp mice resulted further increments in blood pressure and left ventricular hypertrophy, consistent with additive cardiovascular effects. These findings suggest that FGF-23 may participate in unexpected systemic and paracrine networks regulating hemodynamic and myocardial responses.
Collapse
|
42
|
Tian X, Yu C, Shi L, Li D, Chen X, Xia D, Zhou J, Xu W, Ma C, Gu L, An Y. MicroRNA-199a-5p aggravates primary hypertension by damaging vascular endothelial cells through inhibition of autophagy and promotion of apoptosis. Exp Ther Med 2018; 16:595-602. [PMID: 30116316 PMCID: PMC6090226 DOI: 10.3892/etm.2018.6252] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 04/13/2018] [Indexed: 12/19/2022] Open
Abstract
The present study investigated the expression of microRNA (miRNA or miR)-199a-5p in the peripheral blood of patients with primary hypertension, and examined its mechanism of action in vascular endothelial cell injury induced by hypertension. A total of 57 patients with primary hypertension, who were treated at the Affiliated Hospital of Qingdao University (Qingdao, China) between December 2014 and November 2015 were included in the present study. Peripheral blood was collected from all patients. The expression of miR-199a-5p was measured using reverse-transcription quantitative polymerase chain reaction analysis. Human umbilical vein endothelial cells (HUVECs) were divided into negative control, miR-199a-5p mimics and rescue (co-transfected with miR-199a-5p mimics and inhibitor) groups. After transfection, the proliferation and apoptosis of HUVECs were evaluated by a Cell Counting Kit-8 assay, a bromodeoxyuridine incorporation assay and flow cytometry. Western blot analysis was used to determine the expression of proteins involved in autophagy-associated and adenosine monophosphate kinase (AMPK)/unc-51 like autophagy activating kinase 1 (ULK1) signaling pathways. Laser scanning confocal microscopy and electron microscopy were used to observe the autophagy of HUVECs. The expression of miR-199a-5p was elevated in peripheral blood of patients with hypertension, and was correlated with the progression of hypertension. Overexpression of miR-199a-5p inhibited the proliferation and promoted the apoptosis of HUVECs. Upon expression of miR-199a-5p, the transition between microtubule-associated proteins 1A/1B light chain 3B (LC3B)I and LC3BII proteins was inhibited, the expression of p62 protein was upregulated. In addition, miR-199a-5p decreased the numbers of autophagosomes and autolysosomes in HUVECs. The present study demonstrated that expression of miR-199a-5p is positively correlated with the severity of hypertension. Expression of miR-199a-5p aggravated vascular endothelial injury by inhibiting autophagy and promoting the apoptosis of HUVECs via downregulation of the AMPK/ULK1 signaling pathway.
Collapse
Affiliation(s)
- Xintao Tian
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Chunpeng Yu
- Department of Intervention, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Lei Shi
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Dan Li
- Department of Cardiovascular Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Xiaoxue Chen
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Di Xia
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Jingwei Zhou
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Wanqun Xu
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Chengtai Ma
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Lihua Gu
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Yi An
- Department of Cardiovascular Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| |
Collapse
|
43
|
Eguchi S, Kawai T, Scalia R, Rizzo V. Understanding Angiotensin II Type 1 Receptor Signaling in Vascular Pathophysiology. Hypertension 2018; 71:804-810. [PMID: 29581215 DOI: 10.1161/hypertensionaha.118.10266] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Satoru Eguchi
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA.
| | - Tatsuo Kawai
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Rosario Scalia
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Victor Rizzo
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| |
Collapse
|
44
|
Haruhara K, Wakui H, Azushima K, Kurotaki D, Kawase W, Uneda K, Haku S, Kobayashi R, Ohki K, Kinguchi S, Ohsawa M, Minegishi S, Ishigami T, Matsuda M, Yamashita A, Nakajima H, Tamura T, Tsuboi N, Yokoo T, Tamura K. Angiotensin receptor-binding molecule in leukocytes in association with the systemic and leukocyte inflammatory profile. Atherosclerosis 2018; 269:236-244. [PMID: 29407599 DOI: 10.1016/j.atherosclerosis.2018.01.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 12/18/2017] [Accepted: 01/11/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS The components of the renin-angiotensin system in leukocytes is involved in the pathophysiology of non-communicable diseases (NCDs), including hypertension, atherosclerosis and chronic kidney disease. Angiotensin II type 1 receptor (AT1R)-associated protein (ATRAP) is an AT1R-specific binding protein, and is able to inhibit the pathological activation of AT1R signaling in certain animal models of NCDs. The aim of the present study was to investigate the expression and regulation of ATRAP in leukocytes. METHODS Human leukocyte ATRAP mRNA was measured with droplet digital polymerase chain reaction system, and analyzed in relation to the clinical variables. We also examined the leukocyte cytokines mRNA in bone-marrow ATRAP-deficient and wild-type chimeric mice after injection of low-dose lipopolysaccharide. RESULTS The ATRAP mRNA was abundantly expressed in leukocytes, predominantly granulocytes and monocytes, of healthy subjects. In 86 outpatients with NCDs, leukocyte ATRAP mRNA levels correlated positively with granulocyte and monocyte counts and serum C-reactive protein levels. These positive relationships remained significant even after adjustment. Furthermore, the leukocyte ATRAP mRNA was significantly associated with the interleukin-1β, tumor necrosis factor-α and monocyte chemotactic protein-1 mRNA levels in leukocytes of NCDs patients. In addition, the leukocyte interleukin-1β mRNA level was significantly upregulated in bone marrow ATRAP-deficient chimeric mice in comparison to wild-type chimeric mice after injection of lipopolysaccharide. CONCLUSIONS These results suggest that leukocyte ATRAP is an emerging marker capable of reflecting the systemic and leukocyte inflammatory profile, and plays a role as an anti-inflammatory factor in the pathophysiology of NCDs.
Collapse
Affiliation(s)
- Kotaro Haruhara
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore.
| | - Daisuke Kurotaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Wataru Kawase
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazushi Uneda
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sona Haku
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryu Kobayashi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kohji Ohki
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masato Ohsawa
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shintaro Minegishi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoaki Ishigami
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Miyuki Matsuda
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akio Yamashita
- Department of Molecular Biology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hideaki Nakajima
- Department of Hematology and Clinical Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobuo Tsuboi
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| |
Collapse
|
45
|
Abstract
The link between inappropriate salt retention in the kidney and hypertension is well recognized. However, growing evidence suggests that the immune system can play surprising roles in sodium homeostasis, such that the study of inflammatory cells and their secreted effectors has provided important insights into salt sensitivity. As part of the innate immune system, myeloid cells have diverse roles in blood pressure regulation, ranging from prohypertensive actions in the kidney, vasculature, and brain, to effects in the skin that attenuate blood pressure elevation. In parallel, T lymphocyte subsets, as key constituents of the adaptive immune compartment, have variable effects on renal sodium handling and the hypertensive response, accruing from the functions of the cytokines that they produce. Conversely, salt can directly modulate the phenotypes of myeloid and T cells, illustrating bidirectional regulatory mechanisms through which sodium and the immune system coordinately impact blood pressure. This review details the complex interplay between myeloid cells, T cells, and salt in the pathogenesis of essential hypertension.
Collapse
Affiliation(s)
- A Justin Rucker
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Durham Veterans Affairs Medical Center, Durham, North Carolina 27705, USA
| | - Nathan P Rudemiller
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Durham Veterans Affairs Medical Center, Durham, North Carolina 27705, USA
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Durham Veterans Affairs Medical Center, Durham, North Carolina 27705, USA
| |
Collapse
|
46
|
Rodriguez-Iturbe B, Pons H, Johnson RJ. Role of the Immune System in Hypertension. Physiol Rev 2017; 97:1127-1164. [PMID: 28566539 PMCID: PMC6151499 DOI: 10.1152/physrev.00031.2016] [Citation(s) in RCA: 263] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 03/02/2017] [Accepted: 03/02/2017] [Indexed: 02/07/2023] Open
Abstract
High blood pressure is present in more than one billion adults worldwide and is the most important modifiable risk factor of death resulting from cardiovascular disease. While many factors contribute to the pathogenesis of hypertension, a role of the immune system has been firmly established by a large number of investigations from many laboratories around the world. Immunosuppressive drugs and inhibition of individual cytokines prevent or ameliorate experimental hypertension, and studies in genetically-modified mouse strains have demonstrated that lymphocytes are necessary participants in the development of hypertension and in hypertensive organ injury. Furthermore, immune reactivity may be the driving force of hypertension in autoimmune diseases. Infiltration of immune cells, oxidative stress, and stimulation of the intrarenal angiotensin system are induced by activation of the innate and adaptive immunity. High blood pressure results from the combined effects of inflammation-induced impairment in the pressure natriuresis relationship, dysfunctional vascular relaxation, and overactivity of the sympathetic nervous system. Imbalances between proinflammatory effector responses and anti-inflammatory responses of regulatory T cells to a large extent determine the severity of inflammation. Experimental and human studies have uncovered autoantigens (isoketal-modified proteins and heat shock protein 70) of potential clinical relevance. Further investigations on the immune reactivity in hypertension may result in the identification of new strategies for the treatment of the disease.
Collapse
Affiliation(s)
- Bernardo Rodriguez-Iturbe
- Renal Service, Hospital Universitario, Universidad del Zulia, and Instituto Venezolano de Investigaciones Científicas (IVIC)-Zulia, Maracaibo, Venezuela; and Division of Renal Diseases and Hypertension, University of Colorado, Anschutz Campus, Aurora, Colorado
| | - Hector Pons
- Renal Service, Hospital Universitario, Universidad del Zulia, and Instituto Venezolano de Investigaciones Científicas (IVIC)-Zulia, Maracaibo, Venezuela; and Division of Renal Diseases and Hypertension, University of Colorado, Anschutz Campus, Aurora, Colorado
| | - Richard J Johnson
- Renal Service, Hospital Universitario, Universidad del Zulia, and Instituto Venezolano de Investigaciones Científicas (IVIC)-Zulia, Maracaibo, Venezuela; and Division of Renal Diseases and Hypertension, University of Colorado, Anschutz Campus, Aurora, Colorado
| |
Collapse
|
47
|
Wenzel UO, Bode M, Köhl J, Ehmke H. A pathogenic role of complement in arterial hypertension and hypertensive end organ damage. Am J Physiol Heart Circ Physiol 2017; 312:H349-H354. [DOI: 10.1152/ajpheart.00759.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/15/2016] [Accepted: 12/15/2016] [Indexed: 11/22/2022]
Abstract
The self-amplifying cascade of messenger and effector molecules of the complement system serves as a powerful danger-sensing system that protects the host from a hostile microbial environment, while maintaining proper tissue and organ function through effective clearance of altered or dying cells. As an important effector arm of innate immunity, it also plays important roles in the regulation of adaptive immunity. Innate and adaptive immune responses have been identified as crucial players in the pathogenesis of arterial hypertension and hypertensive end organ damage. In line with this view, complement activation may drive the pathology of hypertension and hypertensive injury through its impact on innate and adaptive immune responses. It is well known that complement activation can cause tissue inflammation and injury and complement-inhibitory drugs are effective treatments for several inflammatory diseases. In addition to these proinflammatory properties, complement cleavage fragments of C3 and C5 can exert anti-inflammatory effects that dampen the inflammatory response to injury. Recent experimental data strongly support a role for complement in arterial hypertension. The remarkably similar clinical and histopathological features of malignant nephrosclerosis and atypical hemolytic uremic syndrome, which is driven by complement activation, suggest a role for complement also in the development of malignant nephrosclerosis. Herein, we will review canonical and noncanonical pathways of complement activation as the framework to understand the multiple roles of complement in arterial hypertension and hypertensive end organ damage.
Collapse
Affiliation(s)
- Ulrich O. Wenzel
- Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Marlies Bode
- Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Heimo Ehmke
- Department of Cellular and Integrative Physiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; and
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| |
Collapse
|
48
|
The role of macrophages in hypertension and its complications. Pflugers Arch 2017; 469:419-430. [PMID: 28251313 DOI: 10.1007/s00424-017-1950-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/01/2017] [Accepted: 02/06/2017] [Indexed: 12/17/2022]
Abstract
Circulating monocytes and tissue macrophages play complex roles in the pathogenesis of hypertension, a highly prevalent disease associated with catastrophic cardiovascular morbidity. In the vasculature and kidney, macrophage-derived reactive oxygen species (ROS) and inflammatory cytokines induce endothelial and epithelial dysfunction, respectively, resulting in vascular oxidative stress and impairment of sodium excretion. By contrast, VEGF-C-expressing macrophages in the skin can facilitate the removal of excess interstitial stores of sodium by stimulating lymphangiogenesis. Inappropriate activation of the renin-angiotensin system (RAS) contributes to essential hypertension in a majority of patients, and macrophages express the type 1 (AT1) receptor for angiotensin II (Ang II). While proinflammatory macrophages clearly contribute to RAS-dependent hypertension, activation of the AT1 receptor directly on macrophages suppresses their M1 polarization and limits tubular and interstitial damage to the kidney during hypertension. Thus, stimulating the macrophage AT1 receptor ameliorates the target organ damage and immune stimulation provoked by AT1 receptor activation in intrinsic renal and vascular cells. The proinflammatory cytokines TNF-α and IL-1β produced by M1 macrophages drive blood pressure elevation and consequent target organ damage. However, additional studies are needed to identify the tissues in which these cytokines act and the signaling pathways they stimulate during hypertension. Moreover, identifying the precise myeloid cell subsets that contribute to hypertension should guide the development of more precise immunomodulatory therapies for patients with persistent blood pressure elevation and progressive end-organ injury.
Collapse
|