1
|
Huang S, Zhou Y, Ji H, Zhang T, Liu S, Ma L, Deng D, Ding Y, Han L, Shu S, Wang Y, Chen X. Decoding mechanisms and protein markers in lung-brain axis. Respir Res 2025; 26:190. [PMID: 40390067 PMCID: PMC12090670 DOI: 10.1186/s12931-025-03272-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 05/08/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND The lung-brain axis represents a complex bidirectional communication network that is pivotal in the crosstalk between respiratory and neurological functions. This review summarizes the current understanding of the mechanisms and protein markers that mediate the effects of lung diseases on brain health. MAIN FINDINGS In this review, we explore the mechanisms linking lung injury to neurocognitive impairments, focusing on neural pathways, immune regulation and inflammatory responses, microorganism pathways, and hypoxemia. Specifically, we highlight the role of the vagus nerve in modulating the central nervous system response to pulmonary stimuli; Additionally, the regulatory function of the immune system is underscored, with evidence suggesting that lung-derived immune mediators can traverse the blood-brain barrier, induce neuroinflammation and cognitive decline; Furthermore, we discuss the potential of lung microbiota to influence brain diseases through microbial translocation and immune activation; Finally, the impact of hypoxemia is examined, with findings indicating that it can exacerbate cerebral injury via oxidative stress and impaired perfusion. Moreover, we analyze how pulmonary conditions, such as pneumonia, ALI/ARDS, and asthma, contribute to neurological dysfunction. Prolonged mechanical ventilation can also contribute to cognitive impairment. Conversely, brain diseases (e.g., stroke, traumatic brain injury) can lead to acute respiratory complications. In addition, protein markers such as TLR4, ACE2, A-SAA, HMGB1, and TREM2 are crucial to the lung-brain axis and correlate with disease severity. We also discuss emerging therapeutic strategies targeting this axis, including immunomodulation and microbiome engineering. Overall, understanding the lung-brain interplay is crucial for developing integrated treatment strategies and improving patient outcomes. Further research is needed to elucidate the molecular mechanisms and foster interdisciplinary collaboration.
Collapse
Affiliation(s)
- Shiqian Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Yuxi Zhou
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Haipeng Ji
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Shiya Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Lulin Ma
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Daling Deng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Yuanyuan Ding
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Linlin Han
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Shaofang Shu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Yu Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China.
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China.
| |
Collapse
|
2
|
Príncipe LA, Marchi PH, Cesar CGL, Amaral AR, Duarte KKS, Finardi GLF, Souza JM, Balieiro JCC, Vendramini THA. Evaluation of enzymatically hydrolyzed poultry byproduct meal effects on fecal microbiota and pressure variables in elderly obese cats. Front Vet Sci 2025; 12:1530260. [PMID: 40191086 PMCID: PMC11969457 DOI: 10.3389/fvets.2025.1530260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/17/2025] [Indexed: 04/09/2025] Open
Abstract
Arterial hypertension is influenced by the intestinal microbiota and its metabolites, which play a crucial role in host health. Dietary peptides are multifunctional molecules with therapeutic potential for managing hypertension. This study aimed to evaluate the impact of incorporating enzymatically hydrolyzed poultry byproduct meal (EHPM-c) into extruded dry diets on the fecal microbiota and blood pressure parameters of elderly obese cats. Eighteen owners of neutered, clinically healthy male and female cats of various breeds were randomly assigned to two groups: control (30.8%, conventional poultry byproduct meal-CPM-c) and test (17.07%, CPM-c + 12.0% EHPM-c). Clinical values of systolic blood pressure, serum aldosterone concentrations, angiotensin-converting enzyme I activity, and fecal microbiota using 16S rRNA were measured. Data were processed using SAS software (PROC MIXED, PROC GLIMMIX, and PROC CORR; p < 0.05). Both groups exhibited high microbial alpha diversity, with no significant differences in beta diversity. Although the inclusion of 12.0% EHPM-c had no measurable effect on blood pressure, both diets promoted beneficial modulation of the fecal microbiota, improving intestinal health. These findings underscore the importance of diet in maintaining gut homeostasis in obese senior cats. While the inclusion of 12.0% EHPM-c did not significantly alter blood pressure parameters, the modulation of the fecal microbiota suggests a potential role in maintaining intestinal health. These results highlight the need for further studies to explore different inclusion levels and longer intervention periods.
Collapse
Affiliation(s)
- Leonardo A. Príncipe
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Pedro H. Marchi
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Cinthia G. L. Cesar
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Andressa R. Amaral
- Veterinary Nutrology Service, Veterinary Teaching Hospital, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Kelly K. S. Duarte
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Gabriela L. F. Finardi
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Jennifer M. Souza
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Júlio C. C. Balieiro
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Thiago H. A. Vendramini
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
- Veterinary Nutrology Service, Veterinary Teaching Hospital, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Chhotaray S, Sahoo PK, Mekap SK, Jal S, Pattnaik G. JN. 1 and cardiac-related clinical manifestations: a current public health concern. Front Cardiovasc Med 2024; 11:1488226. [PMID: 39717444 PMCID: PMC11663862 DOI: 10.3389/fcvm.2024.1488226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Affiliation(s)
- Sangeeta Chhotaray
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Bhubaneswar, India
| | - Pralaya Kumar Sahoo
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Bhubaneswar, India
| | - Suman Kumar Mekap
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, India
| | - Soumya Jal
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Bhubaneswar, India
| | - Gurudutta Pattnaik
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, India
| |
Collapse
|
4
|
Kumar VP, Kong Y, Dolland R, Brown SR, Wang K, Dolland D, Mu D, Brown ML. Exploring Angiotensin II and Oxidative Stress in Radiation-Induced Cataract Formation: Potential for Therapeutic Intervention. Antioxidants (Basel) 2024; 13:1207. [PMID: 39456460 PMCID: PMC11504979 DOI: 10.3390/antiox13101207] [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: 09/01/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
Radiation-induced cataracts (RICs) represent a significant public health challenge, particularly impacting individuals exposed to ionizing radiation (IR) through medical treatments, occupational settings, and environmental factors. Effective therapeutic strategies require a deep understanding of the mechanisms underlying RIC formation (RICF). This study investigates the roles of angiotensin II (Ang II) and oxidative stress in RIC development, with a focus on their combined effects on lens transparency and cellular function. Key mechanisms include the generation of reactive oxygen species (ROS) and oxidative damage to lens proteins and lipids, as well as the impact of Ang II on inflammatory responses and cellular apoptosis. While the generation of ROS from water radiolysis is well established, the impact of Ang II on RICs is less understood. Ang II intensifies oxidative stress by activating type 1 receptors (AT1Rs) on lens epithelial cells, resulting in increased ROS production and inflammatory responses. This oxidative damage leads to protein aggregation, lipid peroxidation, and apoptosis, ultimately compromising lens transparency and contributing to cataract formation. Recent studies highlight Ang II's dual role in promoting both oxidative stress and inflammation, which accelerates cataract development. RICs pose a substantial public health concern due to their widespread prevalence and impact on quality of life. Targeting Ang II signaling and oxidative stress simultaneously could represent a promising therapeutic approach. Continued research is necessary to validate these strategies and explore their efficacy in preventing or reversing RIC development.
Collapse
Affiliation(s)
- Vidya P. Kumar
- Armed Forces Radiobiology Research Institute, The Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA;
| | - Yali Kong
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (Y.K.); (K.W.); (D.M.)
| | - Riana Dolland
- Trocar Pharma Inc., 8101 Sandy Spring Rd., Suite 300-W9, Laurel, MD 20707, USA; (R.D.); (D.D.)
| | - Sandra R. Brown
- LensCrafters, Inc., 110 Mall Circle, Suite 2001, Waldorf, MD 20603, USA;
| | - Kan Wang
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (Y.K.); (K.W.); (D.M.)
| | - Damian Dolland
- Trocar Pharma Inc., 8101 Sandy Spring Rd., Suite 300-W9, Laurel, MD 20707, USA; (R.D.); (D.D.)
| | - David Mu
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (Y.K.); (K.W.); (D.M.)
- Leroy T. Canoles, Jr. Cancer Research Center, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Milton L. Brown
- Department of Internal Medicine, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| |
Collapse
|
5
|
Wu Q, Han X, Zhang Y, Liu H, Zhou H, Wang K, Han J. One Copy Number Variation within the Angiopoietin-1 Gene Is Associated with Leizhou Black Goat Meat Quality. Animals (Basel) 2024; 14:2682. [PMID: 39335271 PMCID: PMC11428527 DOI: 10.3390/ani14182682] [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: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
The ANGPT1 gene plays a crucial role in the regulation of angiogenesis and muscle growth, with previous studies identifying copy number variations (CNVs) within this gene among Leizhou black goats. In this study, we investigated three ANGPT1 CNVs in 417 individuals of LZBG using quantitative PCR (qPCR), examining the impact of different CNV types on the ANGPT1 gene expression and their associations with growth and meat quality traits. Notably, the ANGPT1 CNV-1 (ARS1_chr14:24950001-24953600) overlaps with protein-coding regions and conserved domains; its gain-of-copies genotype (copies ≥ 3) was significantly correlated with ANGPT1 mRNA expression in muscle tissue (p < 0.01). Furthermore, the gain-of-copies genotype of CNV-1 demonstrated significant correlations with various phenotypic traits, including carcass weight, body weight, shear stress, chest circumference, and cross-sectional area of longissimus dorsi muscle. These findings indicate that the CNV-1 gain-of-copies genotype in the ANGPT1 gene may serve as a valuable marker for selecting Leizhou black goats exhibiting enhanced growth and muscular development characteristics, thereby holding potential applications in targeted breeding programs aimed at improving meat quality.
Collapse
Affiliation(s)
- Qun Wu
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, No. 5, Shetan Road, Xiashan Area, Zhanjiang 524013, China
| | - Xiaotao Han
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, No. 5, Shetan Road, Xiashan Area, Zhanjiang 524013, China
| | - Yuelang Zhang
- Hainan Institute, Zhejiang University, Sanya 572024, China
| | - Hu Liu
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, No. 5, Shetan Road, Xiashan Area, Zhanjiang 524013, China
| | - Hanlin Zhou
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, No. 5, Shetan Road, Xiashan Area, Zhanjiang 524013, China
| | - Ke Wang
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, No. 5, Shetan Road, Xiashan Area, Zhanjiang 524013, China
| | - Jiancheng Han
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, No. 5, Shetan Road, Xiashan Area, Zhanjiang 524013, China
| |
Collapse
|
6
|
Abstract
The renin-angiotensin system is the most important peptide hormone system in the regulation of cardiovascular homeostasis. Its classical arm consists of the enzymes, renin, and angiotensin-converting enzyme, generating angiotensin II from angiotensinogen, which activates its AT1 receptor, thereby increasing blood pressure, retaining salt and water, and inducing cardiovascular hypertrophy and fibrosis. However, angiotensin II can also activate a second receptor, the AT2 receptor. Moreover, the removal of the C-terminal phenylalanine from angiotensin II by ACE2 (angiotensin-converting enzyme 2) yields angiotensin-(1-7), and this peptide interacts with its receptor Mas. When the aminoterminal Asp of angiotensin-(1-7) is decarboxylated, alamandine is generated, which activates the Mas-related G-protein-coupled receptor D, MrgD (Mas-related G-protein-coupled receptor type D). Since Mas, MrgD, and the AT2 receptor have opposing effects to the classical AT1 receptor, they and the enzymes and peptides activating them are called the alternative or protective arm of the renin-angiotensin system. This review will cover the historical aspects and the current standing of this recent addition to the biology of the renin-angiotensin system.
Collapse
Affiliation(s)
- Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité - University Medicine, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - U. Muscha Steckelings
- Institute for Molecular Medicine, Dept. of Cardiovascular & Renal Research, University of Southern Denmark, Odense, Denmark
| | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Robson A.S. Santos
- National Institute of Science and Technology in Nanobiopharmaceutics (Nanobiofar) - Department of Physiology and Biophysics, Institute of Biological Sciences - Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carlos M. Ferrario
- Laboratory of Translational Hypertension, Department of Surgery, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| |
Collapse
|
7
|
Sabet Sarvestani F, Afshari A, Azarpira N. The role of non-protein-coding RNAs in ischemic acute kidney injury. Front Immunol 2024; 15:1230742. [PMID: 38390339 PMCID: PMC10881863 DOI: 10.3389/fimmu.2024.1230742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Acute kidney injury (AKI) is a condition characterized by a rapid decline in kidney function within a span of 48 hours. It is influenced by various factors including inflammation, oxidative stress, excessive calcium levels within cells, activation of the renin-angiotensin system, and dysfunction in microcirculation. Ischemia-reperfusion injury (IRI) is recognized as a major cause of AKI; however, the precise mechanisms behind this process are not yet fully understood and effective treatments are still needed. To enhance the accuracy of diagnosing AKI during its early stages, the utilization of innovative markers is crucial. Numerous studies suggest that certain noncoding RNAs (ncRNAs), such as long noncoding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), play a central role in regulating gene expression and protein synthesis. These ncRNAs are closely associated with the development and recovery of AKI and have been detected in both kidney tissue and bodily fluids. Furthermore, specific ncRNAs may serve as diagnostic markers and potential targets for therapeutic interventions in AKI. This review aims to summarize the functional roles and changes observed in noncoding RNAs during ischemic AKI, as well as explore their therapeutic potential.
Collapse
Affiliation(s)
| | - Afsoon Afshari
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Batista JPT, de Faria AOV, Ribeiro TFS, Simões e Silva AC. The Role of Renin-Angiotensin System in Diabetic Cardiomyopathy: A Narrative Review. Life (Basel) 2023; 13:1598. [PMID: 37511973 PMCID: PMC10381689 DOI: 10.3390/life13071598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Diabetic cardiomyopathy refers to myocardial dysfunction in type 2 diabetes, but without the traditional cardiovascular risk factors or overt clinical atherosclerosis and valvular disease. The activation of the renin-angiotensin system (RAS), oxidative stress, lipotoxicity, maladaptive immune responses, imbalanced mitochondrial dynamics, impaired myocyte autophagy, increased myocyte apoptosis, and fibrosis contribute to diabetic cardiomyopathy. This review summarizes the studies that address the link between cardiomyopathy and the RAS in humans and presents proposed pathophysiological mechanisms underlying this association. The RAS plays an important role in the development and progression of diabetic cardiomyopathy. The over-activation of the classical RAS axis in diabetes leads to the increased production of angiotensin (Ang) II, angiotensin type 1 receptor activation, and aldosterone release, contributing to increased oxidative stress, fibrosis, and cardiac remodeling. In contrast, Ang-(1-7) suppresses oxidative stress, inhibits tissue fibrosis, and prevents extensive cardiac remodeling. Angiotensin-converting-enzyme (ACE) inhibitors and angiotensin receptor blockers improve heart functioning and reduce the occurrence of diabetic cardiomyopathy. Experimental studies also show beneficial effects for Ang-(1-7) and angiotensin-converting enzyme 2 infusion in improving heart functioning and tissue injury. Further research is necessary to fully understand the pathophysiology of diabetic cardiomyopathy and to translate experimental findings into clinical practice.
Collapse
Affiliation(s)
- João Pedro Thimotheo Batista
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil; (J.P.T.B.); (A.O.V.d.F.); (T.F.S.R.)
| | - André Oliveira Vilela de Faria
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil; (J.P.T.B.); (A.O.V.d.F.); (T.F.S.R.)
| | - Thomas Felipe Silva Ribeiro
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil; (J.P.T.B.); (A.O.V.d.F.); (T.F.S.R.)
| | - Ana Cristina Simões e Silva
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil; (J.P.T.B.); (A.O.V.d.F.); (T.F.S.R.)
- Departamento de Pediatria, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil
| |
Collapse
|
9
|
Askaripour M, Najafipour H, Jafarinejad-Farsangi S, Rajabi S, Saberi S, Jafari E. The effect of daidzein on renal injury in ovariectomized rats: interaction of angiotensin receptors and long non-coding RNAs H19, GAS5, MIAT, and Rian. IRANIAN JOURNAL OF VETERINARY RESEARCH 2023; 24:14-21. [PMID: 37378389 PMCID: PMC10291528 DOI: 10.22099/ijvr.2023.43460.6357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 12/25/2022] [Accepted: 01/01/2023] [Indexed: 06/29/2023]
Abstract
Background Renin-angiotensin system (RAS) is prominently associated with renal pathophysiology in postmenopausal women. Long non-coding RNAs (lncRNAs) H19, GAS5, MIAT, and Rian have been linked to the pathogenesis of renal injury. Aims This study aimed to evaluate the beneficial effects of daidzein on unilateral ureteral obstruction (UUO) induced-renal injury in ovariectomized (OVX) rats through interaction with angiotensin AT1, Mas receptors, and lncRNAs. Methods 84 female rats were ovariectomized (OVX) two weeks before performing obstruction of the left kidney ureter (UUO). The animals were then randomly divided into four main groups (n=21): Sham+DMSO, UUO+DMSO, UUO+17β-Estradiol (E2) (positive control), and UUO+daidzein. Each main group comprised three subgroups (n=7) and were treated with saline, A779 (MasR antagonist), or losartan (AT1R antagonist) for 15 days. On day 16, the animals were euthanized, and the left kidneys were harvested for histopathology and lncRNAs expression assays. Results UUO significantly increased kidney tissue damage score (KTDS) in the UUO rats, increased the expression of H19 and MIAT, and decreased the expression of GAS5 and Rian. Daidzein alone and in co-treatment with losartan or A779 reversed these effects. Daidzein with 1 mg/kg dose was more effective than E2. Conclusion Daidzein alone and in co-treatment with A779 and losartan improved renal injury in UUO rats and recovered dysregulated expression of UUO-related lncRNAs through modulating MasR and AT1R receptors, associating with modulation of the expression of lncRNAs. Daidzein could be considered a renoprotective phytoestrogen substitute for E2 therapy in postmenopausal women suffering from renal diseases.
Collapse
Affiliation(s)
- M Askaripour
- Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
- These authors contributed equally to this work and were considered as the first authors
| | - H Najafipour
- Cardiovascular Research Centre, Institute of Basic and Clinical Physiology Sciences, Afzalipour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | | | - S Rajabi
- Endocrinology and Metabolism Research Centre, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
- These authors contributed equally to this work and were considered as the first authors
| | - Sh Saberi
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
- Physiology Research Centre, Institute of Neuropharmacology, Afzalipour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | - E Jafari
- Pathology and Stem Cell Research Center, Department of Pathology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
10
|
García Martínez JJ, Wozniak H, Salamin P, Giraud R, Le Terrier C, Bendjelid K. Is the prognosis of non-hypertensive, COVID-19 patients treated with renin-angiotensin-aldosterone system inhibitors more uncertain? Physiol Rep 2022; 10:e15512. [PMID: 36397298 PMCID: PMC9672383 DOI: 10.14814/phy2.15512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 06/16/2023] Open
Abstract
Previous studies suggested that ongoing treatment with renin-angiotensin-aldosterone system (RAAS) inhibitor drugs may alter the course of SARS-CoV-2 infection and promote the development of more severe forms of the disease. The authors conducted a comparative, observational study to retrospectively analyze data collected from 394 patients admitted to ICU due to SARS-CoV-2 pneumonia. The primary aim of the study was to establish an association between the use of RAAS inhibitor drugs and mortality in the ICU. The secondary aims of the study were to establish an association between the use of RAAS inhibitor drugs and clinical severity at ICU admission, the need for tracheal intubation, total days of mechanical ventilation, and the ICU length of stay. The authors found no statistically significant difference in ICU mortality between patients on RAAS inhibitor drugs at admission and those who were not (31.3% versus 26.2% mortality, p-value 0.3). However, the group of patients taking RAAS inhibitor drugs appeared to be more critical at ICU admission, and this difference became statistically significant in the subgroup of non-hypertensive patients. ICU mortality in the subgroup of non-hypertensive patients treated with RAAS inhibitor drugs also tended to be higher. Overexpression of the angiotensin-converting enzyme 2 (ACE2) in human cells, induced by RAAS inhibitor drugs, promotes viral entry-replication of SARS-CoV-2 and alters the basal balance of the RAAS, which may explain the findings observed in the present study. These phenomena may be amplified in non-hypertensive patients treated with RAAS inhibitor therapy.
Collapse
Affiliation(s)
- Juan José García Martínez
- Intensive Care UnitHôpital du ValaisSionSwitzerland
- Intensive Care DivisionGeneva University HospitalsGenevaSwitzerland
| | - Hannah Wozniak
- Intensive Care DivisionGeneva University HospitalsGenevaSwitzerland
| | | | - Raphaël Giraud
- Intensive Care DivisionGeneva University HospitalsGenevaSwitzerland
- Geneva Hemodynamic Research GroupGenevaSwitzerland
- Faculty of MedicineGeneva University HospitalsGenevaSwitzerland
| | | | - Karim Bendjelid
- Intensive Care DivisionGeneva University HospitalsGenevaSwitzerland
- Geneva Hemodynamic Research GroupGenevaSwitzerland
- Faculty of MedicineGeneva University HospitalsGenevaSwitzerland
| |
Collapse
|
11
|
Abstract
The coronavirus SARS-CoV-2 infects host cells by binding to the angiotensin-converting enzyme 2 (ACE2) receptor, which belongs to an anti-inflammatory, anti-thrombotic counter-regulatory arm of the renin-angiotensin system (RAS). ACE2 dysfunction and RAS dysregulation has been explored as a driving force in acute respiratory distress syndrome (ARDS), but data from COVID-19 patients has been inconsistent and inconclusive. We sought to identify disruptions of the classical (ACE)/angiotensin (Ang) II/Ang II type-1 receptor (AT1R) and the counter-regulatory ACE2/Ang 1-7/Mas Receptor (MasR) pathways in patients with COVID-19 and correlate these with severity of infection and markers of inflammation and coagulation. Ang II and Ang 1–7 levels in plasma were measured by enzyme-linked immunosorbent assay (ELISA) for 230 patients, 166 of whom were SARS-CoV-2+. Ang 1–7 was repressed in COVID-19 patients compared to that in SARS-CoV-2 negative outpatient controls. Since the control cohort was less sick than the SARS-CoV-2+ group, this association between decreased Ang 1–7 and COVID-19 cannot be attributed to COVID-19 specifically as opposed to critical illness more generally. Multivariable logistic regression analyses demonstrated that every 10-pg/mL increase in plasma Ang 1–7 was associated with a 3% reduction in the odds of hospitalization (adjusted odds ratio [AOR] 0.97, confidence interval [CI] 0.95 to 0.99) and a 3% reduction in odds of requiring oxygen supplementation (AOR 0.97, CI 0.95 to 0.99) and/or ventilation (AOR 0.97, CI 0.94 to 0.99). Ang 1–7 was also inversely associated with pro-inflammatory cytokines and d-dimer in this patient cohort, suggesting that reduced activity in this protective counter-regulatory arm of the RAS contributes to the hyper-immune response and diffuse coagulation activation documented in COVID-19. IMPORTANCE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a unique disease, COVID-19, which ranges in severity from asymptomatic to causing severe respiratory failure and death. Viral transmission throughout the world continues at a high rate despite the development and widespread use of effective vaccines. For those patients who contract COVID-19 and become severely ill, few therapeutic options have been shown to provide benefits and mortality rates are high. Additionally, the pathophysiology underlying COVID-19 disease presentation, progression, and severity is incompletely understood. The significance of our research is in confirming the role of renin-angiotensin system dysfunction in COVID-19 pathogenesis in a large cohort of patients with diverse disease severity and outcomes. Additionally, to our knowledge, this is the first study to pair angiotensin peptide levels with inflammatory and thrombotic markers. These data support the role of ongoing clinical trials examining renin-angiotensin system-targeted therapeutics for the treatment of COVID-19.
Collapse
|
12
|
Bian SZ, Zhang C, Rao RS, Ding XH, Huang L. Systemic Blood Predictors of Elevated Pulmonary Artery Pressure Assessed by Non-invasive Echocardiography After Acute Exposure to High Altitude: A Prospective Cohort Study. Front Cardiovasc Med 2022; 9:866093. [PMID: 35757324 PMCID: PMC9226344 DOI: 10.3389/fcvm.2022.866093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 05/06/2022] [Indexed: 11/14/2022] Open
Abstract
Aim Elevated pulmonary artery pressure (ePAP) in response to high-altitude hypoxia is a critical physiopathological factor in the hypoxic adaptation that may lead to high-altitude pulmonary edema in the acute phase or high-altitude pulmonary hypertension in the long term. However, the sea-level predictors of risk factors for altitude-induced ePAP have not been examined. Thus, we aimed to identify the baseline systemic blood predictors of ePAP after acute high-altitude exposure. Materials and Methods A total of 154 participants were transported to a high altitude 3,700 m from sea level within 2 h. Echocardiography examinations were performed to assess the mean pulmonary artery pressure (mPAP) and hemodynamics at both altitudes. All the individuals underwent blood tests to determine the concentrations of vascular regulatory factors. Univariate and adjusted logistic regression analyses were performed to identify the independent predictors of ePAP and factors related to ePAP. Results The mPAP increased significantly from sea level to high altitude (19.79 ± 6.53–27.16 ± 7.16 mmHg, p < 0.05). Increased levels of endothelin (ET-1), Ang (1–7), Ang II, and bradykinin were found after high-altitude exposure, while the levels of nitric oxide (NO), prostaglandin E2 (PEG2), and serotonin decreased sharply (all p-values < 0.05). At high altitude, 52.6% of the subjects exhibited ePAP, and the mPAP was closely correlated with the baseline Ang II level (r = 0.170, p = 0.036) and follow-up levels of NO (r = −0.209, p = 0.009), Ang II (r = 0.246, p = 0.002), and Ang (1–7) (r = −0.222, p = 0.006) and the left atrial inner diameter (LAD, r = 0.270, p < 0.001). Both the baseline and follow-up NO and Ang II levels were significantly different between the ePAP and non-ePAP groups. Finally, we identified the baseline Ang II and NO concentrations as two independent predictors of ePAP (p < 0.05). We also found that two vascular regulatory factors with inverse roles, namely, Ang (1–7) and Ang II, at high altitudes were independently associated with ePAP. Additionally, ET-1, NO, PEG2, and LAD were associated with ePAP. Conclusion The baseline concentrations of Ang II and NO at sea level are two independent predictors of ePAP after acute high-altitude exposure. Furthermore, Ang (1-7) and Ang II combined with ET-1, NO, PEG2, and LAD at high altitudes may contribute to the development of ePAP.
Collapse
Affiliation(s)
- Shi-Zhu Bian
- Department of Cardiology, Xinqiao Hospital, Institute of Cardiovascular Diseases, Army Medical University (Third Military Medical University), Chongqing, China
| | - Chen Zhang
- Department of Cardiology, Xinqiao Hospital, Institute of Cardiovascular Diseases, Army Medical University (Third Military Medical University), Chongqing, China
| | - Rong-Sheng Rao
- Department of Ultrasonography, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiao-Han Ding
- Department of Health Care and Geriatrics, The 940th Hospital of Joint Logistics Support of Chinese People’s Liberation Army (PLA), Lanzhou, China
| | - Lan Huang
- Department of Cardiology, Xinqiao Hospital, Institute of Cardiovascular Diseases, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Lan Huang,
| |
Collapse
|
13
|
Refined-JinQi-JiangTang tablet ameliorates hypertension through activation of FGF21/FGFR1 axis in fructose-fed rats. J Nat Med 2022; 76:765-773. [PMID: 35534765 DOI: 10.1007/s11418-022-01626-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/20/2022] [Indexed: 10/18/2022]
Abstract
The aim of this study was to investigate the therapeutic effect of JQ-R on metabolic hypertension and its correlation with Fibroblast growth factor 21/Fibroblast growth factor receptors 1(FGF21/FGFR1) pathway. In this study, fructose-induced metabolic hypertension rats were used as hypertension models to detect the regulation effect of JQ-R on hypertension. The effects of JQ-R on blood glucose, blood lipids, serum insulin levels and other metabolic indicators of rats were also measured. The effects of JQ-R on FGF21/FGFR1 signaling pathway in model animals were detected by Real-time quantitative PCR and Western blotting. The results showed that JQ-R significantly reduce the blood pressure of model rats in a dose-dependent manner. Meanwhile, fasting insulin, fasting blood glucose, insulin resistance index, total cholesterol and triglyceride levels were significantly decreased, and glucose and lipid metabolism abnormalities were also significantly improved. JQ-R induces these changes along with FGFR1 phosphorylation, which was also detected in JQ-R treated FGF21 knockout mice. These results suggest that JQ-R can reduce blood pressure and improve glucose and lipid metabolism in fructose-induced hypertension rats. Activation of FGF21/FGFR1 signaling pathway to regulate downstream blood pressure and glucolipid metabolism-related pathways may be one of the important mechanisms of JQ-R in regulating blood pressure.
Collapse
|
14
|
Ferrario CM, Groban L, Wang H, Sun X, VonCannon JL, Wright KN, Ahmad S. The renin–angiotensin system biomolecular cascade: a 2022 update of newer insights and concepts. Kidney Int Suppl (2011) 2022; 12:36-47. [DOI: 10.1016/j.kisu.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/14/2021] [Accepted: 11/08/2021] [Indexed: 12/30/2022] Open
|
15
|
Dias CJ, Costa HA, Alves Dias-Filho CA, Ferreira AC, Rodrigues B, Irigoyen MC, Romão Borges AC, de Andadre Martins V, Branco Vidal FC, Ribeiro RM, Filho NS, Mostarda CT. Carvacrol reduces blood pressure, arterial responsiveness and increases expression of MAS receptors in spontaneously hypertensive rats. Eur J Pharmacol 2022; 917:174717. [PMID: 34953800 DOI: 10.1016/j.ejphar.2021.174717] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/09/2021] [Accepted: 12/20/2021] [Indexed: 12/28/2022]
Abstract
AIM To analyze the effect of the use of carvacrol in the cardiovascular system of spontaneously hypertensive rats (SHR). METHODS Methods: Twenty animals were allocated in four groups, one group control Wistar receiving only sorbitol, used as vehicle of administration of the carvacrol (Wistar-Vehicle), one control group SHR, also receive only sorbitol (SHR-Vehicle), a third, treated with losartan (SHR-Losartan/50 mg/kg), and the fourth, treated with carvacrol (SHR - Carvacrol/20 mg/kg). Sorbitol, losartan and carvacrol were administered by oral gavage daily for 30-day. Hemodynamic variables, vascular reactivity, biochemical parameters, and expression of Mas and AT1 receptors in kidney tissues were analyzed. RESULTS SHR- Carvacrol group showed a maximal effect of inhibition of 56% in the curve of norepinephrine. The Emax of the curves with Ca2+ were smaller in the groups SHR-losartan (40.17%) and SHR-carvacrol (35.71%) when compared to the SHR-Vehicle. The carvacrol increased the expression of the MAS receptors in kidney tissue. CONCLUSION Thirty days of treatment with carvacrol showed an antihypertensive effect associated with less peripheral vascular resistance. Also, treatment with carvacrol increased the expression of MAS receptors in kidney tissue.
Collapse
Affiliation(s)
- Carlos José Dias
- Northeast Biotechnology Network Postgraduate Program (Renorbio), Federal University of Maranhao, St. Luis /Maranhao, Brazil; Cardiovascular Adaptations to Exercise Laboratory (LACORE), Federal University of Maranhão, St. Luis /Maranhao, Brazil; Cardiorenal Adaptations to Exercise Laboratory (LACE), Federal University of Maranhão, Pinheiro /Maranhao, Brazil
| | - Herikson Araújo Costa
- Health Sciences Graduate Program, Federal University of Maranhao, St. Luis /Maranhao, Brazil
| | - Carlos Alberto Alves Dias-Filho
- Northeast Biotechnology Network Postgraduate Program (Renorbio), Federal University of Maranhao, St. Luis /Maranhao, Brazil; Cardiovascular Adaptations to Exercise Laboratory (LACORE), Federal University of Maranhão, St. Luis /Maranhao, Brazil
| | - Andressa Coelho Ferreira
- Cardiovascular Adaptations to Exercise Laboratory (LACORE), Federal University of Maranhão, St. Luis /Maranhao, Brazil; Graduate Program in Adult Health, Federal University of Maranhao, St. Luis /Maranhao, Brazil
| | - Bruno Rodrigues
- Physical Education College, State University of Campinas, Campinas/St Paul, Brazil
| | - Maria Claudia Irigoyen
- Department of Cardiopneumology, Faculty of Medicine of USP, InCor Experimental Hypertension Laboratory, St Paul/St Paul, Brazil
| | - Antônio Carlos Romão Borges
- Northeast Biotechnology Network Postgraduate Program (Renorbio), Federal University of Maranhao, St. Luis /Maranhao, Brazil; Department of Physiological Sciences, Federal University of Maranhao, St. Luis /Maranhao, Brazil
| | - Vicenilma de Andadre Martins
- Northeast Biotechnology Network Postgraduate Program (Renorbio), Federal University of Maranhao, St. Luis /Maranhao, Brazil
| | | | - Rachel Melo Ribeiro
- Department of Physiological Sciences, Federal University of Maranhao, St. Luis /Maranhao, Brazil
| | - Natalino Salgado Filho
- Health Sciences Graduate Program, Federal University of Maranhao, St. Luis /Maranhao, Brazil
| | - Cristiano T Mostarda
- Northeast Biotechnology Network Postgraduate Program (Renorbio), Federal University of Maranhao, St. Luis /Maranhao, Brazil; Cardiovascular Adaptations to Exercise Laboratory (LACORE), Federal University of Maranhão, St. Luis /Maranhao, Brazil; Postgraduate Program in Physical Education, Federal University of Maranhao, St. Luis /Maranhao, Brazil; Graduate Program in Adult Health, Federal University of Maranhao, St. Luis /Maranhao, Brazil.
| |
Collapse
|
16
|
An Adverse Outcomes Approach to Study the Effects of SARS-CoV-2 in 3D Organoid Models. J Mol Biol 2022; 434:167213. [PMID: 34437890 PMCID: PMC8381630 DOI: 10.1016/j.jmb.2021.167213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023]
Abstract
The novel SARS-CoV-2 virus outbreak is the major cause of a respiratory disease known as COVID-19. It has caused a global pandemic and has resulted in mortality in millions. The primary mode of infection is respiratory ailments, however, due to multi-organ complications, COVID-19 patients displays a greater mortality numbers. Due to the 3Rs Principle (Refine, Reduce, Replacement), the scientific community has shifted its focus to 3D organoid models rather than testing animal models. 3D organoid models provide a better physiological architecture as it mimics the real tissue microenvironment and is the best platform to recapitulate organs in a dish. Hence, the organoid approach provides a more realistic drug response in comparison to the traditional 2D cellular models, which lack key physiological relevance due to the absence of proper surface topography and cellular interactions. Furthermore, an adverse outcome pathway (AOPs) provides a best fit model to identify various molecular and cellular events during the exposure of SARS-CoV-2. Hence, 3D organoid research provides information related to gene expression, cell behavior, antiviral studies and ACE2 expression in various organs. In this review, we discuss state-of-the-art lung, liver and kidney 3D organoid system utilizing the AOPs to study SARS-CoV-2 molecular pathogenesis. Furthermore, current challenges are discussed for future application of 3D organoid systems for various disease states.
Collapse
|
17
|
Sakaki Y, Urushihara M, Nagai T, Fujioka K, Kondo S, Kinoshita Y, Hattori T, Kagami S. Urinary angiotensin converting enzyme 2 and disease activity in pediatric IgA nephropathy. THE JOURNAL OF MEDICAL INVESTIGATION 2021; 68:292-296. [PMID: 34759147 DOI: 10.2152/jmi.68.292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Background : Our previous studies demonstrated that the intrarenal renin-angiotensin system (RAS) status was activated in pediatric patients with chronic glomerulonephritis. In the present study, we tested the hypothesis that angiotensin-converting enzyme 2 (ACE2) expression in the kidney is associated with the development of pediatric IgA nephropathy. Methods : We analyzed urinary ACE2 levels and ACE2 expression in the kidney tissues of pediatric patients with IgA nephropathy treated with RAS blockade. Paired tests were used to analyze changes from the first to the second biopsy. Results : Urinary ACE2 levels were significantly decreased after RAS blockade treatment, accompanied by decreased ACE2 expression levels in kidney tissues, urinary protein levels and mesangial hypercellularity scores. Urinary ACE2 levels at the first biopsy were positively correlated with the ACE2 expression levels. Conclusions : These data suggest that urinary ACE2 is associated with ACE2 expression in the diseased kidney, which correlates with the pathogenesis of IgA nephropathy in pediatric patients. J. Med. Invest. 68 : 292-296, August, 2021.
Collapse
Affiliation(s)
- Yuki Sakaki
- Department of Pediatrics, Institute of Biomedical Scicences, The Tokushima University Graduate School, Tokushima, Japan
| | - Maki Urushihara
- Department of Pediatrics, Institute of Biomedical Scicences, The Tokushima University Graduate School, Tokushima, Japan
| | - Takashi Nagai
- Department of Pediatrics, Institute of Biomedical Scicences, The Tokushima University Graduate School, Tokushima, Japan
| | - Keisuke Fujioka
- Department of Pediatrics, Institute of Biomedical Scicences, The Tokushima University Graduate School, Tokushima, Japan
| | - Shuji Kondo
- Department of Pediatrics, Institute of Biomedical Scicences, The Tokushima University Graduate School, Tokushima, Japan
| | - Yukiko Kinoshita
- Department of Pediatrics, Institute of Biomedical Scicences, The Tokushima University Graduate School, Tokushima, Japan
| | - Tomoki Hattori
- Department of Pediatrics, Institute of Biomedical Scicences, The Tokushima University Graduate School, Tokushima, Japan
| | - Shoji Kagami
- Department of Pediatrics, Institute of Biomedical Scicences, The Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
18
|
Elshafei A, Khidr EG, El-Husseiny AA, Gomaa MH. RAAS, ACE2 and COVID-19; a mechanistic review. Saudi J Biol Sci 2021; 28:6465-6470. [PMID: 34305426 PMCID: PMC8270731 DOI: 10.1016/j.sjbs.2021.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/26/2021] [Accepted: 07/04/2021] [Indexed: 01/08/2023] Open
Abstract
The use of angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) in coronavirus disease 2019 (COVID-19) patients has been claimed as associated with the risk of COVID-19 infection and its subsequent morbidities and mortalities. These claims were resulting from the possibility of upregulating the expression of angiotensin-converting enzyme 2 (ACE2), facilitation of SARS-CoV-2 entry, and increasing the susceptibility of infection in such treated cardiovascular patients. ACE2 and renin-angiotensin-aldosterone system (RAAS) products have a critical function in controlling the severity of lung injury, fibrosis, and failure following the initiation of the disease. This review is to clarify the mechanisms beyond the possible deleterious effects of angiotensin II (Ang II), and the potential protective role of angiotensin 1-7 (Ang 1-7) against pulmonary fibrosis, with a subsequent discussion of the latest updates on ACEIs/ARBs use and COVID-19 susceptibility in the light of these mechanisms and biochemical explanation.
Collapse
Key Words
- ACE1, angiotensin-converting enzyme 1
- ACE2
- ACE2, angiotensin-converting enzyme 2
- ACEIs
- ACEIs, angiotensin-converting enzyme inhibitors
- AEC-II, alveolar epithelial type II cells
- ARBs
- ARBs, angiotensin receptor blockers
- AT1R, angiotensin type 1 receptor
- AT2R, angiotensin type 2 receptor
- Ang 1-7, angiotensin 1-7
- Ang 1-9, angiotensin 1-9
- AngI, angiotensin I
- AngII, angiotensin II
- Angiotensin 1–7
- Angiotensin II
- COVID-19
- COVID-19, coronavirus disease 2019
- CVD, cardiovascular disease
- ERK, extracellular signal-regulated kinase
- ICU, intensive care unit
- MAPK, mitogen-activated protein kinase
- NLRP3, (NOD, LRR, and pyrin domain-containing protein 3)
- RAAS, renin-angiotensin-aldosterone system
- TGF-β, transforming growth factor-beta
- miR-21, microRNA-21
Collapse
Affiliation(s)
- Ahmed Elshafei
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Emad Gamil Khidr
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed A. El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Maher H. Gomaa
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| |
Collapse
|
19
|
Formanowicz D, Gutowska K, Szawulak B, Formanowicz P. The Crosstalk between SARS-CoV-2 Infection and the RAA System in Essential Hypertension-Analyses Using Systems Approach. Int J Mol Sci 2021; 22:ijms221910518. [PMID: 34638859 PMCID: PMC8508810 DOI: 10.3390/ijms221910518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/19/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), responsible for the coronavirus disease of 2019 (COVID-19) pandemic, has affected and continues to affect millions of people across the world. Patients with essential arterial hypertension and renal complications are at particular risk of the fatal course of this infection. In our study, we have modeled the selected processes in a patient with essential hypertension and chronic kidney disease (CKD) suffering from COVID-19, emphasizing the function of the renin-angiotensin-aldosterone (RAA) system. The model has been built in the language of Petri nets theory. Using the systems approach, we have analyzed how COVID-19 may affect the studied organism, and we have checked whether the administration of selected anti-hypertensive drugs (angiotensin-converting enzyme inhibitors (ACEIs) and/or angiotensin receptor blockers (ARBs)) may impact the severity of the infection. Besides, we have assessed whether these drugs effectively lower blood pressure in the case of SARS-CoV-2 infection affecting essential hypertensive patients. Our research has shown that neither the ACEIs nor the ARBs worsens the course infection. However, when assessing the treatment of hypertension in the active SARS-CoV-2 infection, we have observed that ARBs might not effectively reduce blood pressure; they may even have the slightly opposite effect. On the other hand, we have confirmed the effectiveness of arterial hypertension treatment in patients receiving ACEIs. Moreover, we have found that the simultaneous use of ARBs and ACEIs averages the effects of taking both drugs, thus leading to only a slight decrease in blood pressure. We are a way from suggesting that ARBs in all hypertensive patients with COVID-19 are ineffective, but we have shown that research in this area should still be continued.
Collapse
Affiliation(s)
- Dorota Formanowicz
- Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
| | - Kaja Gutowska
- Institute of Computing Science, Poznan University of Technology, 60-965 Poznan, Poland; (K.G.); (B.S.)
| | - Bartłomiej Szawulak
- Institute of Computing Science, Poznan University of Technology, 60-965 Poznan, Poland; (K.G.); (B.S.)
| | - Piotr Formanowicz
- Institute of Computing Science, Poznan University of Technology, 60-965 Poznan, Poland; (K.G.); (B.S.)
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland
- Correspondence:
| |
Collapse
|
20
|
Commentary on: Renin-angiotensin system overactivation in perivascular adipose tissue contributes to vascular dysfunction in heart failure. Clin Sci (Lond) 2021; 135:683-686. [PMID: 33649765 DOI: 10.1042/cs20210017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 11/17/2022]
Abstract
We comment on the publication of a paper in which Brazilian investigators evaluate the anticontractile response of perivascular adipose tissue (PVAT) in experimental heart failure (HF) induced in rats by occlusion of a coronary artery.
Collapse
|
21
|
Drugs repurposing against SARS-CoV2 and the new variant B.1.1.7 (alpha strain) targeting the spike protein: molecular docking and simulation studies. Heliyon 2021; 7:e07803. [PMID: 34423145 PMCID: PMC8367657 DOI: 10.1016/j.heliyon.2021.e07803] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/20/2021] [Accepted: 08/12/2021] [Indexed: 12/23/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) is responsible for the global COVID-19 pandemic and millions of deaths worldwide. In December 2020, a new alpha strain of SARS-CoV2 was identified in the United Kingdom. It was referred to as VUI 202012/01 (Alpha strain modelled under investigation, 2020, month 12, number 01). The interaction between spike protein and ACE2 receptor is a prerequisite for entering virion into the host cell. The present study is focussed on the spike protein of the SARS-COV 2, involving the comparison of binding affinity of new alpha strain modelled spike with previous strain spike (PDB ID:7DDN) using in silico molecular docking, dynamics and simulation studies. The molecular docking studies of the alpha strain modelled spike protein confirmed its higher affinity for the ACE2 receptor than the spike protein of the dominant strain. Similar computational approaches have also been used to investigate the potency of FDA approved drugs from the ZINC Database against the spike protein of new alpha strain modelled and old ones. The drug molecules which showed strong affinity for both the spike proteins are then subjected to ADME analysis. The overall binding energy of Conivaptan (-107.503 kJ/mol) and Trosec (-94.029 kJ/mol) is indicative of their strong binding affinities, well supported by interactions with critical residues. We investigated the potential FDA drugs for repurposing against the spike protein of alpha strain modelled of SARS-CoV-2. Spike protein of alpha strain modelled of SARS-CoV-2 has more affinity for the ACE2 receptor of host cell than previous strains and, therefore, is more contagious. Conivaptan, Ecamsule and Trosec are common drugs that bind strongly with spike protein of both the strains . Molecular Docking and simulation studies show that Conivaptan and Trosec emerged as potential inhibitors of the alpha strain modelled spike protein.
Collapse
|
22
|
de Souza AMA, Ecelbarger CM, Sandberg K. Caloric Restriction and Cardiovascular Health: the Good, the Bad, and the Renin-Angiotensin System. Physiology (Bethesda) 2021; 36:220-234. [PMID: 34159807 DOI: 10.1152/physiol.00002.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Much excitement exists over the cardioprotective and life-extending effects of caloric restriction (CR). This review integrates population studies with experimental animal research to address the positive and negative impact of mild and severe CR on cardiovascular physiology and pathophysiology, with a particular focus on the renin-angiotensin system (RAS). We also highlight the gaps in knowledge and areas ripe for future physiological research.
Collapse
Affiliation(s)
- Aline M A de Souza
- Division of Nephrology and Hypertension, Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Carolyn M Ecelbarger
- Division of Nephrology and Hypertension, Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Kathryn Sandberg
- Division of Nephrology and Hypertension, Department of Medicine, Georgetown University, Washington, District of Columbia
| |
Collapse
|
23
|
Imatinib (STI571) Inhibits the Expression of Angiotensin-Converting Enzyme 2 and Cell Entry of the SARS-CoV-2-Derived Pseudotyped Viral Particles. Int J Mol Sci 2021; 22:ijms22136938. [PMID: 34203261 PMCID: PMC8268654 DOI: 10.3390/ijms22136938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 11/17/2022] Open
Abstract
A group of clinically approved cancer therapeutic tyrosine kinase inhibitors was screened to test their effects on the expression of angiotensin-converting enzyme 2 (ACE2), the cell surface receptor for SARS-CoV-2. Here, we show that the receptor tyrosine kinase inhibitor imatinib (also known as STI571, Gleevec) can inhibit the expression of the endogenous ACE2 gene at both the transcript and protein levels. Treatment with imatinib resulted in inhibition of cell entry of the viral pseudoparticles (Vpps) in cell culture. In FVB mice orally fed imatinib, tissue expression of ACE2 was reduced, specifically in the lungs and renal tubules, but not in the parenchyma of other organs such as the heart and intestine. Our finding suggests that receptor tyrosine kinases play a role in COVID-19 infection and can be therapeutic targets with combined treatments of the best conventional care of COVID-19.
Collapse
|
24
|
Behl T, Shah S, Kaur I, Yadav S, Kanwar R, Seth S, Wig N, Sharma KK, Yadav HN. Role of ACE 2 and Vitamin D: The Two Players in Global Fight against COVID-19 Pandemic. ANNALS OF THE NATIONAL ACADEMY OF MEDICAL SCIENCES (INDIA) 2021. [DOI: 10.1055/s-0041-1729781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AbstractThe global pandemic of coronavirus disease 2019 (COVID-19) has spread across the borders, gaining attention from both health care professional and researchers to understand the mode of entry and actions induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), its causative agent in the human body. The role of angiotensin-converting enzyme–2 (ACE2) in facilitating the entry of the virus in the host cell by binding to it is similar to SARS-CoV-1, the causative agent for severe acute respiratory syndrome (SARS) which emerged in 2003. Besides the role of ACE2 as a molecular target for the virus, the review displays the potential benefits of ACE2 enzyme and various agents that modify its activity in curbing the effects of the deadly virus, thus unfolding a dual character of ACE2 in the current pandemic. As evident by the differences in the susceptibility toward viral infection in children and geriatric population, it must be noted that the older population has limited ACE2 levels and greater infection risk, whereas the situation is reversed in the case of the pediatric population, demonstrating the defensive character of ACE2 in the latter, despite acting as receptor target for SARS-CoV-2. Also, the upregulation of ACE2 levels by estrogen has indicated greater resistance to infection in females than in the male human population. ACE2 is a carboxypeptidase, which degrades angiotensin II and counteracts its actions to protect against cardiovascular risks associated with the virus. Another contribution of this enzyme is supported by the role of circulating soluble ACE2, which acts as a receptor to bind the virus but does not mediate its actions, therefore blocking its interaction to membrane-bound ACE2 receptors. The review also shares the enhanced risks of developing COVID-19 infection by using ACE inhibitors and ARBs. However, both these agents have been reported to upregulate ACE2 levels; yet, adequate evidence regarding their role is quite inconsistent in human studies. Furthermore, the role of vitamin D has been highlighted in regulating the immune system of the body through renin-angiotensin-aldosterone system (RAAS) inhibition, by downregulating host cell receptor expression to prevent virus attachment. Besides, vitamin D also acts through several other mechanisms like upregulating antimicrobial peptides, fighting against the proinflammatory milieu created by the invading virus, and interfering with the viral replication cycle as well as calcitriol-mediated blockage of CREB protein. Hypovitaminosis D is attributed to elevated risks of acute respiratory distress syndrome (ARDS), lung damage, and cardiovascular disorders, further increasing the severity of COVID-19 infection.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara University, Punjab, India
| | - Sadia Shah
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Ishnoor Kaur
- Department of Pharmacology, Chitkara University, Punjab, India
| | - Sushma Yadav
- Department of Obstetrics and Gynaecology, Shaheed Hasan Khan Mewati Government Medical College, Haryana, India
| | - Raj Kanwar
- Department of Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep Seth
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Naveet Wig
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - K. K. Sharma
- National Academy of Medical Sciences (India), New Delhi, India
| | | |
Collapse
|
25
|
Gholami M, Safari S, Ulloa L, Motaghinejad M. Neuropathies and neurological dysfunction induced by coronaviruses. J Neurovirol 2021; 27:380-396. [PMID: 33983506 PMCID: PMC8117458 DOI: 10.1007/s13365-021-00977-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/23/2021] [Accepted: 04/05/2021] [Indexed: 02/03/2023]
Abstract
During the recent years, viral epidemic due to coronaviruses, such as SARS (Severe Acute Respiratory Syndrome), Middle East Respiratory Coronavirus Syndrome (MERS), and COVID-19 (coronavirus disese-19), has become a global problem. In addition to causing cardiovascular and respiratory lethal dysfunction, these viruses can cause neurodegeneration leading to neurological disorders. Review of the current scientific literature reveals the multiple neuropathies and neuronal dysfunction associated with these viruses. Here, we review the major findings of these studies and discuss the main neurological sequels and outcomes of coronavirus infections with SARS, MERS, and COVID-19. This article analyzes and discusses the main mechanisms of coronavirus-induced neurodegeneration according to the current experimental and clinical studies. Coronaviruses can damage the nerves directly through endovascular dysfunctions thereby affecting nerve structures and synaptic connections. Coronaviruses can also induce neural cell degeneration indirectly via mitochondrial dysfunction inducing oxidative stress, inflammation, and apoptosis. Thus, coronaviruses can cause neurological disorders by inducing neurovascular dysfunction affecting nerve structures and synaptic connections, and by inducing inflammation, oxidative stress, and apoptosis. While some of these mechanisms are similar to other RNA viruses, the neurotoxic mechanisms of COVID-19, MERS, and SARS-CoV viruses are unknown and need detailed clinical and experimental studies.
Collapse
Affiliation(s)
- Mina Gholami
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Safari
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, NC, 27710, Durham, USA.
| | - Majid Motaghinejad
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Dysregulation of the Renin-Angiotensin-Aldosterone System (RAA) in Patients Infected with SARS-CoV-2-Possible Clinical Consequences. Int J Mol Sci 2021; 22:ijms22094503. [PMID: 33925881 PMCID: PMC8123500 DOI: 10.3390/ijms22094503] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 01/22/2023] Open
Abstract
SARS-CoV-2 impairs the renin-angiotensin-aledosterone system via binding ACE2 enzyme. ACE2 plays a key role in the biosynthesis of angiotensin (1-7), catalyzing the conversion of angiotensin 2 into angiotensin (1-7) and the reaction of angiotensin synthesis (1-9), from which angiotensin is (1-7) produced under the influence of ACE (Angiotensin-Converting Enzyme). Angiotensin 2 is a potent vasoconstrictor and atherogenic molecule converted by ACE2 to reducing inflammation and vasodilating in action angiotensin (1-7). Angiotensin (1-9), that is a product of angiotensin 1 metabolism and precursor of angiotensin (1-7), also exerts cell protective properties. Balance between angiotensin 2 and angiotensin (1-7) regulates blood pressure and ACE2 plays a critical role in this balance. ACE2, unlike ACE, is not inhibited by ACE inhibitors at the doses used in humans during the treatment of arterial hypertension. Membrane ACE2 is one of the receptors that allows SARS-CoV-2 to enter the host cells. ACE2 after SARS-CoV-2 binding is internalized and degraded. Hence ACE2 activity on the cell surface is reduced leading to increase the concentration of angiotensin 2 and decrease the concentration of angiotensin (1-7). Disturbed angiotensins metabolism, changes in ratio between angiotensins with distinct biological activities leading to domination of atherogenic angiotensin 2 can increase the damage to the lungs.
Collapse
|
27
|
Wysocki J, Ye M, Hassler L, Gupta AK, Wang Y, Nicoleascu V, Randall G, Wertheim JA, Batlle D. A Novel Soluble ACE2 Variant with Prolonged Duration of Action Neutralizes SARS-CoV-2 Infection in Human Kidney Organoids. J Am Soc Nephrol 2021; 32:795-803. [PMID: 33526471 PMCID: PMC8017551 DOI: 10.1681/asn.2020101537] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND There is an urgent need for approaches to prevent and treat SARS-CoV-2 infection. Administration of soluble ACE2 protein acting as a decoy to bind to SARS-CoV-2 should limit viral uptake mediated by binding to membrane-bound full-length ACE2, and further therapeutic benefit should result from ensuring enzymatic ACE2 activity to affected organs in patients with COVID-19. METHODS A short variant of human soluble ACE2 protein consisting of 618 amino acids (hACE2 1-618) was generated and fused with an albumin binding domain (ABD) using an artificial gene encoding ABDCon, with improved albumin binding affinity. Human kidney organoids were used for infectivity studies of SARS-CoV-2 in a BSL-3 facility to examine the neutralizing effect of these novel ACE2 variants. RESULTS Whereas plasma ACE2 activity of the naked ACE2 1-618 and ACE2 1-740 lasted about 8 hours, the ACE2 1-618-ABD resulted in substantial activity at 96 hours, and it was still biologically active 3 days after injection. Human kidney organoids express ACE2 and TMPRSS2, and when infected with SARS-CoV-2, our modified long-acting ACE2 variant neutralized infection. CONCLUSIONS This novel ACE2 1-618-ABD can neutralize SARS-CoV-2 infectivity in human kidney organoids, and its prolonged duration of action should ensure improved efficacy to prevent viral escape and dosing convenience.
Collapse
Affiliation(s)
- Jan Wysocki
- Division of Nephrology and Hypertension, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Minghao Ye
- Division of Nephrology and Hypertension, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Luise Hassler
- Division of Nephrology and Hypertension, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ashwani Kumar Gupta
- Comprehensive Transplant Center, Department of Surgery, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Yuguo Wang
- Comprehensive Transplant Center, Department of Surgery, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Vlad Nicoleascu
- Department of Microbiology, Ricketts Laboratory, University of Chicago, Chicago, Illinois
| | - Glenn Randall
- Department of Microbiology, Ricketts Laboratory, University of Chicago, Chicago, Illinois
| | - Jason A. Wertheim
- Comprehensive Transplant Center, Department of Surgery, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Daniel Batlle
- Division of Nephrology and Hypertension, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
28
|
Eshaq RS, Watts MN, Carter PR, Leskova W, Aw TY, Alexander JS, Harris NR. Candesartan Normalizes Changes in Retinal Blood Flow and p22phox in the Diabetic Rat Retina. PATHOPHYSIOLOGY 2021; 28:86-97. [PMID: 35366272 PMCID: PMC8830460 DOI: 10.3390/pathophysiology28010008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 11/23/2022] Open
Abstract
Angiotensin II has been implicated in the progression of diabetic retinopathy, which is characterized by altered microvasculature, oxidative stress, and neuronal dysfunction. The signaling induced by angiotensin II can occur not only via receptor-mediated calcium release that causes vascular constriction, but also through a pathway whereby angiotensin II activates NADPH oxidase to elicit the formation of reactive oxygen species (ROS). In the current study, we administered the angiotensin II receptor antagonist candesartan (or vehicle, in untreated animals) in a rat model of type 1 diabetes in which hyperglycemia was induced by injection of streptozotocin (STZ). Eight weeks after the STZ injection, untreated diabetic rats were found to have a significant increase in tissue levels of angiotensin converting enzyme (ACE; p < 0.05) compared to non-diabetic controls, a 33% decrease in retinal blood flow rate (p < 0.001), and a dramatic increase in p22phox (a subunit of the NADPH oxidase). The decrease in retinal blood flow, and the increases in retinal ACE and p22phox in the diabetic rats, were all significantly attenuated (p < 0.05) by the administration of candesartan in drinking water within one week. Neither STZ nor candesartan induced any changes in tissue levels of superoxide dismutase (SOD-1), 4-hydroxynonenal (4-HNE), or nitrotyrosine. We conclude that one additional benefit of candesartan (and other angiotensin II antagonists) may be to normalize retinal blood flow, which may have clinical benefits in diabetic retinopathy.
Collapse
|
29
|
Batchu SN, Kaur H, Yerra VG, Advani SL, Kabir MG, Liu Y, Klein T, Advani A. Lung and Kidney ACE2 and TMPRSS2 in Renin-Angiotensin System Blocker-Treated Comorbid Diabetic Mice Mimicking Host Factors That Have Been Linked to Severe COVID-19. Diabetes 2021; 70:759-771. [PMID: 33310740 DOI: 10.2337/db20-0765] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/27/2020] [Indexed: 12/20/2022]
Abstract
The causes of the increased risk of severe coronavirus disease 2019 (COVID-19) in people with diabetes are unclear. It has been speculated that renin-angiotensin system (RAS) blockers may promote COVID-19 by increasing ACE2, which severe acute respiratory syndrome coronavirus 2 uses to enter host cells, along with the host protease TMPRSS2. Taking a reverse translational approach and by combining in situ hybridization, primary cell isolation, immunoblotting, quantitative RT-PCR, and liquid chromatography-tandem mass spectrometry, we studied lung and kidney ACE2 and TMPRSS2 in diabetic mice mimicking host factors linked to severe COVID-19. In healthy young mice, neither the ACE inhibitor ramipril nor the AT1 receptor blocker telmisartan affected lung or kidney ACE2 or TMPRSS2, except for a small increase in kidney ACE2 protein with ramipril. In contrast, mice with comorbid diabetes (aging, high-fat diet, and streptozotocin-induced diabetes) had heightened lung ACE2 and TMPRSS2 protein levels and increased lung ACE2 activity. None of these parameters were affected by RAS blockade. ACE2 was similarly upregulated in the kidneys of mice with comorbid diabetes compared with aged controls, whereas TMPRSS2 (primarily distal nephron) was highest in telmisartan-treated animals. Upregulation of lung ACE2 activity in comorbid diabetes may contribute to an increased risk of severe COVID-19. This upregulation is driven by comorbidity and not by RAS blockade.
Collapse
Affiliation(s)
- Sri Nagarjun Batchu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Harmandeep Kaur
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Veera Ganesh Yerra
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Suzanne L Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - M Golam Kabir
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Youan Liu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Thomas Klein
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma, Biberach, Germany
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Chen IC, Lin JY, Liu YC, Chai CY, Yeh JL, Hsu JH, Wu BN, Dai ZK. Angiotensin-Converting Enzyme 2 Activator Ameliorates Severe Pulmonary Hypertension in a Rat Model of Left Pneumonectomy Combined With VEGF Inhibition. Front Med (Lausanne) 2021; 8:619133. [PMID: 33681251 PMCID: PMC7933511 DOI: 10.3389/fmed.2021.619133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/25/2021] [Indexed: 11/28/2022] Open
Abstract
Background: Pulmonary arterial hypertension (PAH) is a life-threatening and deteriorating disease with no promising therapy available currently due to its diversity and complexity. An imbalance between vasoconstriction and vasodilation has been proposed as the mechanism of PAH. Angiotensin-converting enzyme 2 (ACE2), which catalyzes the hydrolysis of the vasoconstrictor angiotensin (Ang) II into the vasodilator Ang-(1-7), has been shown to be an important regulator of blood pressure and cardiovascular diseases. Herein we hypothesized diminazene aceturate (DIZE), an ACE2 activator, could ameliorate the development of PAH and pulmonary vascular remodeling. Methods: A murine model of PAH was established using left pneumonectomy (PNx) on day 0 followed by injection of a single dose of the VEGF receptor-2 inhibitor SU5416 (25 mg/kg) subcutaneously on day 1. All hemodynamic and biochemical measurements were done at the end of the study on day 42. Animals were divided into 4 groups (n = 6–8/group): (1) sham-operated group, (2) vehicle-treatment group (SuPNx42), (3) early treatment group (SuPNx42/DIZE1−42) with DIZE at 15 mg/kg/day, subcutaneously from day 1 to day 42, and (4) late treatment group (SuPNx42/DIZE29−42) with DIZE from days 29–42. Results: In both the early and late treatment groups, DIZE significantly attenuated the mean pulmonary artery pressure, pulmonary arteriolar remodeling, and right ventricle brain natriuretic peptide (BNP), as well as reversed the overexpression of ACE while up-regulating the expression of Ang-(1-7) when compared with the vehicle-treatment group. In addition, the early treatment group also significantly decreased plasma BNP and increased the expression of eNOS. Conclusions: ACE2 activator has therapeutic potentials for preventing and attenuating the development of PAH in an animal model of left pneumonectomy combined with VEGF inhibition. Activation of ACE2 may thus be a useful therapeutic strategy for the treatment of human PAH.
Collapse
Affiliation(s)
- I-Chen Chen
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,College of Medicine, Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jao-Yu Lin
- Department of Surgery, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ching Liu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jwu-Lai Yeh
- College of Medicine, Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jong-Hau Hsu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bin-Nan Wu
- College of Medicine, Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zen-Kong Dai
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,College of Medicine, Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
31
|
Gul R, Kim UH, Alfadda AA. Renin-angiotensin system at the interface of COVID-19 infection. Eur J Pharmacol 2021; 890:173656. [PMID: 33086029 PMCID: PMC7568848 DOI: 10.1016/j.ejphar.2020.173656] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/07/2020] [Accepted: 10/16/2020] [Indexed: 11/30/2022]
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been recognized as a potential entry receptor for SARS-CoV-2 infection. Binding of SARS-CoV-2 to ACE2 allows engagement with pulmonary epithelial cells and pulmonary infection with the virus. ACE2 is an essential component of renin-angiotensin system (RAS), and involved in promoting protective effects to counter-regulate angiotensin (Ang) II-induced pathogenesis. The use of angiotensin receptor blockers (ARBs) and ACE inhibitors (ACEIs) was implicitly negated during the early phase of COVID-19 pandemic, considering the role of these antihypertensive agents in enhancing ACE2 expression thereby promoting the susceptibility to SARS-CoV-2. However, no clinical data has supported this assumption, but indeed evidence demonstrates that ACEIs and ARBs, besides their cardioprotective effects in COVID-19 patients with cardiovascular diseases, might also be beneficial in acute lung injuries by preserving the ACE2 function and switching the balance from deleterious ACE/Ang II/AT1 receptor axis towards a protective ACE2/Ang (1-7)/Mas receptor axis.
Collapse
Affiliation(s)
- Rukhsana Gul
- Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Saudi Arabia.
| | - Uh-Hyun Kim
- Department of Biochemistry & National Creative Research Laboratory for Ca(2+) Signaling, Chonbuk National University Medical School, Jeonju, 54907, Republic of Korea
| | - Assim A Alfadda
- Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Saudi Arabia; Department of Medicine, College of Medicine, King Saud University, PO Box 2925, Riyadh, 11461, Saudi Arabia; Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
32
|
Marquez A, Wysocki J, Pandit J, Batlle D. An update on ACE2 amplification and its therapeutic potential. Acta Physiol (Oxf) 2021; 231:e13513. [PMID: 32469114 PMCID: PMC7267104 DOI: 10.1111/apha.13513] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/11/2022]
Abstract
The renin angiotensin system (RAS) plays an important role in the pathogenesis of variety of diseases. Targeting the formation and action of angiotensin II (Ang II), the main RAS peptide, has been the key therapeutic target for last three decades. ACE‐related carboxypeptidase (ACE2), a monocarboxypeptidase that had been discovered 20 years ago, is one of the catalytically most potent enzymes known to degrade Ang II to Ang‐(1‐7), a peptide that is increasingly accepted to have organ‐protective properties that oppose and counterbalance those of Ang II. In addition to its role as a RAS enzyme ACE2 is the main receptor for SARS‐CoV‐2. In this review, we discuss various strategies that have been used to achieve amplification of ACE2 activity including the potential therapeutic potential of soluble recombinant ACE2 protein and novel shorter ACE2 variants.
Collapse
Affiliation(s)
- Alonso Marquez
- Feinberg Medical SchoolNorthwestern University Chicago IL USA
- Department of Medicine Division of Nephrology and Hypertension Chicago IL USA
| | - Jan Wysocki
- Feinberg Medical SchoolNorthwestern University Chicago IL USA
- Department of Medicine Division of Nephrology and Hypertension Chicago IL USA
| | - Jay Pandit
- Feinberg Medical SchoolNorthwestern University Chicago IL USA
- Department of Medicine Division of Nephrology and Hypertension Chicago IL USA
| | - Daniel Batlle
- Feinberg Medical SchoolNorthwestern University Chicago IL USA
- Department of Medicine Division of Nephrology and Hypertension Chicago IL USA
| |
Collapse
|
33
|
Positive association of angiotensin II receptor blockers, not angiotensin-converting enzyme inhibitors, with an increased vulnerability to SARS-CoV-2 infection in patients hospitalized for suspected COVID-19 pneumonia. PLoS One 2020; 15:e0244349. [PMID: 33347477 PMCID: PMC7751849 DOI: 10.1371/journal.pone.0244349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/08/2020] [Indexed: 01/19/2023] Open
Abstract
Background Angiotensin-converting enzyme 2 is the receptor that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses for entry into lung cells. Because ACE-2 may be modulated by angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin II receptor blockers (ARBs), there is concern that patients treated with ACEIs and ARBs are at higher risk of coronavirus disease 2019 (COVID-19) pneumonia. Aim This study sought to analyze the association of COVID-19 pneumonia with previous treatment with ACEIs and ARBs. Materials and methods We retrospectively reviewed 684 consecutive patients hospitalized for suspected COVID-19 pneumonia and tested by polymerase chain reaction assay. Patients were split into two groups, according to whether (group 1, n = 484) or not (group 2, n = 250) COVID-19 was confirmed. Multivariable adjusted comparisons included a propensity score analysis. Results The mean age was 63.6 ± 18.7 years, and 302 patients (44%) were female. Hypertension was present in 42.6% and 38.4% of patients in groups 1 and 2, respectively (P = 0.28). Treatment with ARBs was more frequent in group 1 than group 2 (20.7% vs. 12.0%, respectively; odds ratio [OR] 1.92, 95% confidence interval [CI] 1.23–2.98; P = 0.004). No difference was found for treatment with ACEIs (12.7% vs. 15.7%, respectively; OR 0.81, 95% CI 0.52–1.26; P = 0.35). Propensity score-matched multivariable logistic regression confirmed a significant association between COVID-19 and previous treatment with ARBs (adjusted OR 2.36, 95% CI 1.38–4.04; P = 0.002). Significant interaction between ARBs and ACEIs for the risk of COVID-19 was observed in patients aged > 60 years, women, and hypertensive patients. Conclusions This study suggests that ACEIs and ARBs are not similarly associated with COVID-19. In this retrospective series, patients with COVID-19 pneumonia more frequently had previous treatment with ARBs compared with patients without COVID-19.
Collapse
|
34
|
Singh TU, Parida S, Lingaraju MC, Kesavan M, Kumar D, Singh RK. Drug repurposing approach to fight COVID-19. Pharmacol Rep 2020; 72:1479-1508. [PMID: 32889701 PMCID: PMC7474498 DOI: 10.1007/s43440-020-00155-6] [Citation(s) in RCA: 285] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023]
Abstract
Currently, there are no treatment options available for the deadly contagious disease, coronavirus disease 2019 (COVID-19). Drug repurposing is a process of identifying new uses for approved or investigational drugs and it is considered as a very effective strategy for drug discovery as it involves less time and cost to find a therapeutic agent in comparison to the de novo drug discovery process. The present review will focus on the repurposing efficacy of the currently used drugs against COVID-19 and their mechanisms of action, pharmacokinetics, dosing, safety, and their future perspective. Relevant articles with experimental studies conducted in-silico, in-vitro, in-vivo, clinical trials in humans, case reports, and news archives were selected for the review. Number of drugs such as remdesivir, favipiravir, ribavirin, lopinavir, ritonavir, darunavir, arbidol, chloroquine, hydroxychloroquine, tocilizumab and interferons have shown inhibitory effects against the SARS-CoV2 in-vitro as well as in clinical conditions. These drugs either act through virus-related targets such as RNA genome, polypeptide packing and uptake pathways or target host-related pathways involving angiotensin-converting enzyme-2 (ACE2) receptors and inflammatory pathways. Using the basic knowledge of viral pathogenesis and pharmacodynamics of drugs as well as using computational tools, many drugs are currently in pipeline to be repurposed. In the current scenario, repositioning of the drugs could be considered the new avenue for the treatment of COVID-19.
Collapse
Affiliation(s)
- Thakur Uttam Singh
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, Uttar Pradesh, India.
| | - Subhashree Parida
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, Uttar Pradesh, India
| | - Madhu Cholenahalli Lingaraju
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, Uttar Pradesh, India
| | - Manickam Kesavan
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, Uttar Pradesh, India
| | - Dinesh Kumar
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, Uttar Pradesh, India
| | - Raj Kumar Singh
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, Uttar Pradesh, India
| |
Collapse
|
35
|
Sifat AE, Nozohouri S, Villalba H, Vaidya B, Abbruscato TJ. The Role of Smoking and Nicotine in the Transmission and Pathogenesis of COVID-19. J Pharmacol Exp Ther 2020; 375:498-509. [PMID: 33033170 PMCID: PMC7718727 DOI: 10.1124/jpet.120.000170] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/30/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 virus, is turning out to be one of the most devastating global pandemics in the history of humankind. There is a shortage of effective therapeutic strategies or preventative vaccines for this disease to date. A rigorous investigation is needed for identifying and developing more effective therapeutic strategies for COVID-19. Angiotensin-converting enzyme 2 (ACE2), a crucial factor in COVID-19 pathogenesis, has been identified as a potential target for COVID-19 treatment. Smoking and vaping are potential risk factors for COVID-19 that are also shown to upregulate ACE2 expression. In this review, we have discussed the pathobiology of COVID-19 in the lungs and brain and the role of ACE2 in the transmission and pathobiology of this disease. Furthermore, we have shown possible interactions between nicotine/smoking and ACE2 in the lungs and brain, which could aggravate the transmission and pathobiology of COVID-19, resulting in a poor disease outcome. SIGNIFICANCE STATEMENT: This review addresses the present global pandemic of coronavirus disease 2019 (COVID-19) with respect to its pathobiology in the lungs and brain. It focuses on the potential negative impact of tobacco and nicotine exposure on the outcomes of this disease by interaction with the angiotensin-converting enzyme 2 receptor. It adds to the time-sensitive and critically important growing knowledge about the risk factors, transmission, pathobiology, and prognosis of COVID-19.
Collapse
Affiliation(s)
- Ali Ehsan Sifat
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Heidi Villalba
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| |
Collapse
|
36
|
Davidson AM, Wysocki J, Batlle D. Interaction of SARS-CoV-2 and Other Coronavirus With ACE (Angiotensin-Converting Enzyme)-2 as Their Main Receptor: Therapeutic Implications. Hypertension 2020; 76:1339-1349. [PMID: 32851855 PMCID: PMC7480804 DOI: 10.1161/hypertensionaha.120.15256] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 originated from Wuhan, China, in December 2019 and rapidly spread to other areas worldwide. Since then, coronavirus disease 2019 (COVID-19) has reached pandemic proportions with >570 000 deaths globally by mid-July 2020. The magnitude of the outbreak and the potentially severe clinical course of COVID-19 has led to a burst of scientific research on this novel coronavirus and its host receptor ACE (angiotensin-converting enzyme)-2. ACE2 is a homolog of the ACE that acts on several substrates in the renin-Ang (angiotensin) system. With unprecedented speed, scientific research has solved the structure of SARS-CoV-2 and imaged its binding with the ACE2 receptor. In SARS-CoV-2 infection, the viral S (spike) protein receptor-binding domain binds to ACE2 to enter the host cell. ACE2 expression in the lungs is relatively low, but it is present in type II pneumocytes-a cell type also endowed with TMPRSS2 (transmembrane protease serine 2). This protease is critical for priming the SARS-CoV-2 S protein to complex with ACE2 and enter the cells. Herein, we review the current understanding of the interaction of SARS-CoV-2 with ACE2 as it has rapidly unfolded over the last months. While it should not be assumed that we have a complete picture of SARS-CoV-2 mechanism of infection and its interaction with ACE2, much has been learned with clear therapeutic implications. Potential therapies aimed at intercepting SARS-CoV-2 from reaching the full-length membrane-bound ACE2 receptor using soluble ACE2 protein and other potential approaches are briefly discussed as well.
Collapse
Affiliation(s)
- Anne M. Davidson
- From the Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Jan Wysocki
- From the Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Daniel Batlle
- From the Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
37
|
Georges JL, Cochet H, Roger G, Ben Jemaa H, Soltani J, Azowa JB, Mamou R, Gilles F, Saba J, Prevot A, Pasqualini M, Monguillon V, De Tournemire M, Bertrand A, Koukabi-Fradelizi M, Beressi JP, Livarek B. [Association of hypertension and antihypertensive agents and the severity of COVID-19 pneumonia. A monocentric French prospective study]. Ann Cardiol Angeiol (Paris) 2020; 69:247-254. [PMID: 33039120 PMCID: PMC7522617 DOI: 10.1016/j.ancard.2020.09.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM Angiotensin converting enzyme (ACE) type 2 is the receptor of SARSCoV-2 for cell entry into lung cells. Because ACE-2 may be modulated by ACE inhibitors (ACEIs) and angiotensin II receptor blockers (ARBs), there are concern that patients treated with ACEIs and ARBs are at higher risk for COVID-19 infection or severity. This study sought to analyse the association of severe forms of COVID-19 and mortality with hypertension and a previous treatment with ACEI and ARB. METHODS Prospective follow-up of 433 consecutive patients hospitalised for COVID-19 pneumonia confirmed by PCR or highly probable on clinical, biological, and radiological findings, and included in the COVHYP study. Mortality and severe COVID-19 (criteria: death, intensive care unit, or hospitalisation >30 days) were compared in patients receiving or not ACEIs and ARBs. Follow-up was 100% at hospital discharge, and 96.5% at >1month. RESULTS Age was 63.6±18.7 years, and 40%) were female. At follow-up (mean 78±50 days), 136 (31%) patients had severity criteria (death, 64 ; intensive care unit, 73; hospital stay >30 days, 49). Hypertension (55.1% vs 36.7%, P<0.001) and antihypertensive treatment were associated with severe COVID-19 and mortality. The association between ACEI/ARB treatment and COVID-19 severity criteria found in univariate analysis (Odds Ratio 1.74, 95%CI [1.14-2.64], P=0.01) was not confirmed when adjusted on age, gender, and hypertension (adjusted OR1.13 [0.59-2.15], P=0.72). Diabetes and hypothyroidism were associated with severe COVID-19, whereas history of asthma was not. CONCLUSION This study suggests that previous treatment with ACEI and ARB is not associated with hospital mortality, 1- and 2-month mortality, and severity criteria in patients hospitalised for COVID-19. No protective effect of ACEIs and ARBs on severe pneumonia related to COVID-19 was demonstrated.
Collapse
Affiliation(s)
- J-L Georges
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France.
| | - H Cochet
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - G Roger
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - H Ben Jemaa
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - J Soltani
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - J-B Azowa
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - R Mamou
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - F Gilles
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - J Saba
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - A Prevot
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - M Pasqualini
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - V Monguillon
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - M De Tournemire
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - A Bertrand
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - M Koukabi-Fradelizi
- Service d'accueil des urgences, Centre Hospitalier de Versailles, Le Chesnay, France
| | - J-P Beressi
- Service de diabétologie, Centre Hospitalier de Versailles, Le Chesnay, France
| | - B Livarek
- Service de cardiologie, Centre Hospitalier de Versailles, Le Chesnay, France
| |
Collapse
|
38
|
Ferrario CM, Ahmad S, Groban L. Twenty years of progress in angiotensin converting enzyme 2 and its link to SARS-CoV-2 disease. Clin Sci (Lond) 2020; 134:2645-2664. [PMID: 33063823 PMCID: PMC9055624 DOI: 10.1042/cs20200901] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 12/22/2022]
Abstract
The virulence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the aggressive nature of the disease has transformed the universal pace of research in the desperate attempt to seek effective therapies to halt the morbidity and mortality of this pandemic. The rapid sequencing of the SARS-CoV-2 virus facilitated identification of the receptor for angiotensin converting enzyme 2 (ACE2) as the high affinity binding site that allows virus endocytosis. Parallel evidence that coronavirus disease 2019 (COVID-19) disease evolution shows greater lethality in patients with antecedent cardiovascular disease, diabetes, or even obesity questioned the potential unfavorable contribution of angiotensin converting enzyme (ACE) inhibitors or angiotensin II (Ang II) receptor blockers as facilitators of adverse outcomes due to the ability of these therapies to augment the transcription of Ace2 with consequent increase in protein formation and enzymatic activity. We review, here, the specific studies that support a role of these agents in altering the expression and activity of ACE2 and underscore that the robustness of the experimental data is associated with weak clinical long-term studies of the existence of a similar regulation of tissue or plasma ACE2 in human subjects.
Collapse
Affiliation(s)
- Carlos M. Ferrario
- Departments of Surgery and Physiology-Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, U.S.A
| | - Sarfaraz Ahmad
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, U.S.A
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC 27157, U.S.A
| |
Collapse
|
39
|
Jaiswal G, Kumar V. In-silico design of a potential inhibitor of SARS-CoV-2 S protein. PLoS One 2020; 15:e0240004. [PMID: 33002032 PMCID: PMC7529220 DOI: 10.1371/journal.pone.0240004] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/18/2020] [Indexed: 12/25/2022] Open
Abstract
The SARS-CoV-2 virus has caused a pandemic and is public health emergency of international concern. As of now, no registered therapies are available for treatment of coronavirus infection. The viral infection depends on the attachment of spike (S) glycoprotein to human cell receptor angiotensin-converting enzyme 2 (ACE2). We have designed a protein inhibitor (ΔABP-D25Y) targeting S protein using computational approach. The inhibitor consists of two α helical peptides homologues to protease domain (PD) of ACE2. Docking studies and molecular dynamic simulation revealed that the inhibitor binds exclusively at the ACE2 binding site of S protein. The computed binding affinity of the inhibitor is higher than the ACE2 and thus will likely out compete ACE2 for binding to S protein. Hence, the proposed inhibitor ΔABP-D25Y could be a potential blocker of S protein and receptor binding domain (RBD) attachment.
Collapse
Affiliation(s)
- Grijesh Jaiswal
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida Uttar Pradesh, India
| | - Veerendra Kumar
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida Uttar Pradesh, India
| |
Collapse
|
40
|
Abstract
Purpose of Review Angiotensin-converting enzyme 2 (ACE2), a specific high-affinity angiotensin II-hydrolytic enzyme, is the vector that facilitates cellular entry of SARS-CoV-1 and the novel SARS-CoV-2 coronavirus. SARS-CoV-2, which crossed species barriers to infect humans, is highly contagious and associated with high lethality due to multi-organ failure, mostly in older patients with other co-morbidities. Recent Findings Accumulating clinical evidence demonstrates that the intensity of the infection and its complications are more prominent in men. It has been postulated that potential functional modulation of ACE2 by estrogen may explain the sex difference in morbidity and mortality. Summary We review here the evidence regarding the role of estrogenic hormones in ACE2 expression and regulation, with the intent of bringing to the forefront potential mechanisms that may explain sex differences in SARS-CoV-2 infection and COVID-19 outcomes, assist in management of COVID-19, and uncover new therapeutic strategies.
Collapse
|
41
|
Assadi F. Why Children are Less Likely to Contract COVID-19 Infection than Adults? Int J Prev Med 2020; 11:74. [PMID: 32742618 PMCID: PMC7373085 DOI: 10.4103/ijpvm.ijpvm_199_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 04/18/2020] [Indexed: 11/04/2022] Open
Affiliation(s)
- Farahnak Assadi
- Department of Pediatrics, Division of Nephrology, Rush University Medical Sciences, Chicago, Illinois, USA
| |
Collapse
|
42
|
Holappa M, Vapaatalo H, Vaajanen A. Local ocular renin-angiotensin-aldosterone system: any connection with intraocular pressure? A comprehensive review. Ann Med 2020; 52:191-206. [PMID: 32308046 PMCID: PMC7877937 DOI: 10.1080/07853890.2020.1758341] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/15/2020] [Indexed: 12/28/2022] Open
Abstract
The renin-angiotensin system (RAS) is one of the oldest and most extensively studied human peptide cascades, well-known for its role in regulating blood pressure. When aldosterone is included, RAAS is involved also in fluid and electrolyte homeostasis. There are two main axes of RAAS: (1) Angiotensin (1-7), angiotensin converting enzyme 2 and Mas receptor (ACE2-Ang(1-7)-MasR), (2) Angiotensin II, angiotensin converting enzyme 1 and angiotensin II type 1 receptor (ACE1-AngII-AT1R). In its entirety, RAAS comprises dozens of angiotensin peptides, peptidases and seven receptors. The first mentioned axis is known to counterbalance the deleterious effects of the latter axis. In addition to the systemic RAAS, tissue-specific regulatory systems have been described in various organs, evidence that RAAS is both an endocrine and an autocrine system. These local regulatory systems, such as the one present in the vascular endothelium, are responsible for long-term regional changes. A local RAAS and its components have been detected in many structures of the human eye. This review focuses on the local ocular RAAS in the anterior part of the eye, its possible role in aqueous humour dynamics and intraocular pressure as well as RAAS as a potential target for anti-glaucomatous drugs.KEY MESSAGESComponents of renin-angiotensin-aldosterone system have been detected in different structures of the human eye, introducing the concept of a local intraocular renin-angiotensin-aldosterone system (RAAS).Evidence is accumulating that the local ocular RAAS is involved in aqueous humour dynamics, regulation of intraocular pressure, neuroprotection and ocular pathology making components of RAAS attractive candidates when developing new effective ways to treat glaucoma.
Collapse
Affiliation(s)
- Mervi Holappa
- Medical Faculty, Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Heikki Vapaatalo
- Medical Faculty, Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Anu Vaajanen
- Department of Ophthalmology, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
43
|
|
44
|
Palmeira P, Barbuto JAM, Silva CAA, Carneiro-Sampaio M. Why is SARS-CoV-2 infection milder among children? Clinics (Sao Paulo) 2020; 75:e1947. [PMID: 32428111 PMCID: PMC7213663 DOI: 10.6061/clinics/2020/e1947] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Affiliation(s)
- Patricia Palmeira
- Laboratorio de Investigacao Medica (LIM-36), Departamento de Pediatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - José Alexandre M Barbuto
- Departamento Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Clovis Artur A Silva
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Magda Carneiro-Sampaio
- Laboratorio de Investigacao Medica (LIM-36), Departamento de Pediatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
45
|
Miesbach W. Pathological Role of Angiotensin II in Severe COVID-19. TH OPEN 2020; 4:e138-e144. [PMID: 32607467 PMCID: PMC7319800 DOI: 10.1055/s-0040-1713678] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/19/2020] [Indexed: 01/08/2023] Open
Abstract
The activated renin-angiotensin system induces a prothrombotic state resulting from the imbalance between coagulation and fibrinolysis. Angiotensin II is the central effector molecule of the activated renin-angiotensin system and is degraded by the angiotensin-converting enzyme 2 to angiotensin (1-7). The novel coronavirus infection (classified as COVID-19) is caused by the new coronavirus SARS-CoV-2 and is characterized by an exaggerated inflammatory response that can lead to severe manifestations such as acute respiratory distress syndrome, sepsis, and death in a proportion of patients, mostly elderly patients with preexisting comorbidities. SARS-CoV-2 uses the angiotensin-converting enzyme 2 receptor to enter the target cells, resulting in activation of the renin-angiotensin system. After downregulating the angiotensin-converting enzyme 2, the vasoconstrictor angiotensin II is increasingly produced and its counterregulating molecules angiotensin (1-7) reduced. Angiotensin II increases thrombin formation and impairs fibrinolysis. Elevated levels were strongly associated with viral load and lung injury in patients with severe COVID-19. Therefore, the complex clinical picture of patients with severe complications of COVID-19 is triggered by the various effects of highly expressed angiotensin II on vasculopathy, coagulopathy, and inflammation. Future treatment options should focus on blocking the thrombogenic and inflammatory properties of angiotensin II in COVID-19 patients.
Collapse
Affiliation(s)
- Wolfgang Miesbach
- Department of Haemostaseology and Haemophilia Center, Institute of Transfusion Medicine, Medical Clinic 2, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
46
|
The Vasoactive Mas Receptor in Essential Hypertension. J Clin Med 2020; 9:jcm9010267. [PMID: 31963731 PMCID: PMC7019597 DOI: 10.3390/jcm9010267] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/17/2022] Open
Abstract
The renin–angiotensin–aldosterone system (RAAS) has been studied extensively, and with the inclusion of novel components, it has become evident that the system is much more complex than originally anticipated. According to current knowledge, there are two main axes of the RAAS, which counteract each other in terms of vascular control: The classical vasoconstrictive axis, renin/angiotensin-converting enzyme/angiotensin II/angiotensin II receptor type 1 (AT1R), and the opposing vasorelaxant axis, angiotensin-converting enzyme 2/angiotensin-(1-7)/Mas receptor (MasR). An abnormal activity within the system constitutes a hallmark in hypertension, which is a global health problem that predisposes cardiovascular and renal morbidities. In particular, essential hypertension predominates in the hypertensive population of more than 1.3 billion humans worldwide, and yet, the pathophysiology behind this multifactorial condition needs clarification. While commonly applied pharmacological strategies target the classical axis of the RAAS, discovery of the vasoprotective effects of the opposing, vasorelaxant axis has presented encouraging experimental evidence for a new potential direction in RAAS-targeted therapy based on the G protein-coupled MasR. In addition, the endogenous MasR agonist angiotensin-(1-7), peptide analogues, and related molecules have become the subject of recent studies within this field. Nevertheless, the clinical potential of MasR remains unclear due to indications of physiological-biased activities of the RAAS and interacting signaling pathways.
Collapse
|
47
|
ACE-Triggered Hypertension Incites Stroke: Genetic, Molecular, and Therapeutic Aspects. Neuromolecular Med 2019; 22:194-209. [PMID: 31802381 DOI: 10.1007/s12017-019-08583-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/29/2019] [Indexed: 01/13/2023]
Abstract
Stroke is the second largest cause of death worldwide. Angiotensin converting enzyme (ACE) gene has emerged as an important player in the pathogenesis of hypertension and consequently stroke. It encodes ACE enzyme that converts the inactive decapeptide angiotensin I to active octapeptide, angiotensin II (Ang II). Dysregulation in the expression of ACE gene, on account of genetic variants or regulation by miRNAs, alters the levels of ACE in the circulation. Variable expression of ACE affects the levels of Ang II. Ang II acts through different signal transduction pathways via various tyrosine kinases (receptor/non-receptor) and protein serine/threonine kinases, initiating a downstream cascade of molecular events. In turn these activated molecular pathways might lead to hypertension and inflammation thereby resulting in cardiovascular and cerebrovascular diseases including stroke. In order to regulate the overexpression of ACE, many ACE inhibitors and blockers have been developed, some of which are still under clinical trials.
Collapse
|
48
|
Rajapaksha IG, Gunarathne LS, Asadi K, Cunningham SC, Sharland A, Alexander IE, Angus PW, Herath CB. Liver-Targeted Angiotensin Converting Enzyme 2 Therapy Inhibits Chronic Biliary Fibrosis in Multiple Drug-Resistant Gene 2-Knockout Mice. Hepatol Commun 2019; 3:1656-1673. [PMID: 31832573 PMCID: PMC6887688 DOI: 10.1002/hep4.1434] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 09/07/2019] [Indexed: 12/11/2022] Open
Abstract
There is a large unmet need for effective therapies for cholestatic disorders, including primary sclerosing cholangitis (PSC), a disease that commonly results in liver failure. Angiotensin (Ang) II of the renin Ang system (RAS) is a potent profibrotic peptide, and Ang converting enzyme 2 (ACE2) of the alternate RAS breaks down Ang II to antifibrotic peptide Ang‐(1‐7). In the present study, we investigated long‐term effects of ACE2 delivered by an adeno‐associated viral vector and short‐term effects of Ang‐(1‐7) peptide in multiple drug‐resistant gene 2‐knockout (Mdr2‐KO) mice. These mice develop progressive biliary fibrosis with pathologic features closely resembling those observed in PSC. A single intraperitoneal injection of ACE2 therapy markedly reduced liver injury (P < 0.05) and biliary fibrosis (P < 0.01) at both established (3‐6 months of age) and advanced (7‐9 months of age) disease compared to control vector‐injected Mdr2‐KO mice. This was accompanied by increased hepatic Ang‐(1‐7) levels (P < 0.05) with concomitant reduction in hepatic Ang II levels (P < 0.05) compared to controls. Moreover, Ang‐(1‐7) peptide infusion improved liver injury (P < 0.05) and biliary fibrosis (P < 0.0001) compared to saline‐infused disease controls. The therapeutic effects of both ACE2 therapy and Ang‐(1‐7) infusion were associated with significant (P < 0.01) reduction in hepatic stellate cell (HSC) activation and collagen expression. While ACE2 therapy prevented the loss of epithelial characteristics of hepatocytes and/or cholangiocytes in vivo, Ang‐(1‐7) prevented transdifferentiation of human cholangiocytes (H69 cells) into the collagen‐secreting myofibroblastic phenotype in vitro. We showed that an increased ratio of hepatic Ang‐(1‐7) to Ang II levels by ACE2 therapy results in the inhibition of HSC activation and biliary fibrosis. Conclusion: ACE2 therapy has the potential to treat patients with biliary diseases, such as PSC.
Collapse
Affiliation(s)
- Indu G Rajapaksha
- Department of Medicine University of Melbourne Austin Health Heidelberg Australia
| | - Lakmie S Gunarathne
- Department of Medicine University of Melbourne Austin Health Heidelberg Australia
| | | | - Sharon C Cunningham
- Children's Medical Research Institute School of Medicine University of Sydney Sydney Australia
| | - Alexandra Sharland
- Central Clinical School School of Medicine University of Sydney Sydney Australia
| | - Ian E Alexander
- Children's Medical Research Institute School of Medicine University of Sydney Sydney Australia
| | - Peter W Angus
- Department of Medicine University of Melbourne Austin Health Heidelberg Australia
| | - Chandana B Herath
- Department of Medicine University of Melbourne Austin Health Heidelberg Australia
| |
Collapse
|
49
|
Shao Z, Schuster A, Borowski AG, Thakur A, Li L, Wilson Tang WH. Soluble angiotensin converting enzyme 2 levels in chronic heart failure is associated with decreased exercise capacity and increased oxidative stress-mediated endothelial dysfunction. Transl Res 2019; 212:80-88. [PMID: 31323221 PMCID: PMC6755052 DOI: 10.1016/j.trsl.2019.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 05/28/2019] [Accepted: 06/30/2019] [Indexed: 01/08/2023]
Abstract
The aim of this study was to explore the relationship between serum soluble angiotensin converting enzyme 2 (sACE2), parameters of cardiopulmonary exercise testing and plasma asymmetric dimethylarginine (ADMA), a marker of oxidative stress-induced endothelial dysfunction. This has not been previously evaluated. We assessed 50 consecutive ambulatory patients with chronic systolic heart failure and left ventricular ejection fraction (LVEF) ≤45%. Their blood samples were collected for sACE2 and ADMA tests before they underwent symptom-limited cardiopulmonary exercise testing and transthoracic echocardiography. The majority of our study subjects had New York Heart Association functional class II (74%) and III (18%) presentation, and 42% of patients had ischemic etiology. Median sACE2 activity was 10.36 (7.00-14.47) ng/mL and mean ADMA was 0.90 ± 0.22. sACE2 activity was inversely correlated with pVO2 (r = -0.42, P = 0.00283), exercise time (r = -0.35, P = 0.0138) and LVEF (r = -0.548, P < 0.001), and positively correlated with VE/VCO2 slope (r = 0.294, P = 0.0405), ΔDBP (r = 0.315, P = 0.0278), mitral E/Ea ratio (r = 0.442, P = 0.00158) and ADMA levels (r = 0.351, P = 0.0134). Meanwhile, we observed a negative correlation between ADMA and pVO2 (r = -0.424, P = 0.00227) and positive correlations between ADMA and VE/VCO2 slope (r = 0.515, P < 0.001), ΔDBP (r = 0.391, P = 0.00568), mitral E/Ea ratio (r = 0.426, P = 0.00219). In multivariate logistic regression analysis, sACE2 was independently associated with peak oxygen uptake (% predicted) after adjusting for body mass index (BMI) and mitral E/Ea ratio (odds ratio [OR] 0.81 (0.58-0.94), P = 0.041) and associated with oxygen pulse (VO2/HR) (%) after adjusting for age, gender, BMI and mitral E/Ea ratio (OR 0.83 [0.68-0.95], P = 0.025). Therefore in stable chronic systolic heart failure patients, higher sACE2 activity is independently associated with diminished exercise capacity and correlates with elevated systemic oxidative stress-mediated endothelial dysfunction.
Collapse
Affiliation(s)
- Zhili Shao
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Allen G Borowski
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio
| | - Akanksha Thakur
- Department of Medicine, Stanford University Medical Center, Stanford, California
| | - Lin Li
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Wai Hong Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
50
|
Differential Gene Expression Profile of Renin-Angiotensin System in the Left Atrium in Mitral Regurgitation Patients. DISEASE MARKERS 2018; 2018:6924608. [PMID: 30581499 PMCID: PMC6276386 DOI: 10.1155/2018/6924608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 11/01/2018] [Indexed: 11/17/2022]
Abstract
Background Left atrial enlargement is a mortality and heart failure risk factor in primary mitral regurgitation (MR) patients. Pig models of MR have shown differential expression of genes linked to the renin-angiotensin system. Therefore, the aim of this study was to investigate the key genes of the renin-angiotensin that are expressed differentially in the left atrial myocardium in MR patients. Methods Quantitative RT-PCR was used to compare gene expression in the renin-angiotensin system in the left atrium in MR patients, aortic valve disease patients, and normal subjects. Results Plasma angiotensin II concentrations did not significantly differ between MR patients and aortic valve disease patients (P = 0.582). Compared to normal controls, however, MR patients had significantly downregulated expressions of angiotensin-converting enzyme, angiotensin I converting enzyme 2, type 1 angiotensin II receptor, glutamyl aminopeptidase, angiotensinogen, cathepsin A (CTSA), thimet oligopeptidase 1, neurolysin, alanyl aminopeptidase, cathepsin G, leucyl/cystinyl aminopeptidase (LNPEP), neprilysin, and carboxypeptidase A3 in the left atrium. The MR patients also had significantly upregulated expressions of MAS1 oncogene (MAS1) and mineralocorticoid receptor compared to normal controls. Additionally, in comparison with aortic valve disease patients, MR patients had significantly downregulated CTSA and LNPEP expression and significantly upregulated MAS1 expression in the left atrium. Conclusions Expressions of genes in the renin-angiotensin system, especially CTSA, LNPEP, and MAS1, in the left atrium in MR patients significantly differed from expressions of these genes in aortic valve disease patients and normal controls. Notably, differences in expression were independent of circulating angiotensin II levels. The results of this study provide a rationale for pharmacological therapies or posttranslational regulation therapies targeting genes expressed differentially in the renin-angiotensin system to remedy structural remodeling associated with atrial enlargement and heart failure progression in patients with MR.
Collapse
|