101
|
Karmouty-Quintana H, Thandavarayan RA, Keller SP, Sahay S, Pandit LM, Akkanti B. Emerging Mechanisms of Pulmonary Vasoconstriction in SARS-CoV-2-Induced Acute Respiratory Distress Syndrome (ARDS) and Potential Therapeutic Targets. Int J Mol Sci 2020; 21:E8081. [PMID: 33138181 PMCID: PMC7662604 DOI: 10.3390/ijms21218081] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
The 1918 influenza killed approximately 50 million people in a few short years, and now, the world is facing another pandemic. In December 2019, a novel coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused an international outbreak of a respiratory illness termed coronavirus disease 2019 (COVID-19) and rapidly spread to cause the worst pandemic since 1918. Recent clinical reports highlight an atypical presentation of acute respiratory distress syndrome (ARDS) in COVID-19 patients characterized by severe hypoxemia, an imbalance of the renin-angiotensin system, an increase in thrombogenic processes, and a cytokine release storm. These processes not only exacerbate lung injury but can also promote pulmonary vascular remodeling and vasoconstriction, which are hallmarks of pulmonary hypertension (PH). PH is a complication of ARDS that has received little attention; thus, we hypothesize that PH in COVID-19-induced ARDS represents an important target for disease amelioration. The mechanisms that can promote PH following SARS-CoV-2 infection are described. In this review article, we outline emerging mechanisms of pulmonary vascular dysfunction and outline potential treatment options that have been clinically tested.
Collapse
Affiliation(s)
- Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Divisions of Pulmonary, Critical Care and Sleep Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | | | - Steven P. Keller
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Sandeep Sahay
- Co-Director, Pulmonary Vascular Diseases Center, The Methodist Hospital, Houston, TX 77030, USA;
| | - Lavannya M. Pandit
- Michael E. DeBakey Veterans Affairs Medical Center, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Bindu Akkanti
- Divisions of Pulmonary, Critical Care and Sleep Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| |
Collapse
|
102
|
Gao Y, Liu T, Zhong W, Liu R, Zhou H, Huang W, Zhang W. Risk of Metformin in Patients With Type 2 Diabetes With COVID-19: A Preliminary Retrospective Report. Clin Transl Sci 2020; 13:1055-1059. [PMID: 32955785 PMCID: PMC7537216 DOI: 10.1111/cts.12897] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/12/2020] [Indexed: 01/08/2023] Open
Abstract
The current outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has spread across the world. No specific antiviral agents have been adequately evidenced for the treatment of coronavirus disease 2019 (COVID-19). Although metformin has been recommended as a host-directed therapy for COVID-19, there are some opposite views. The effects of metformin on the disease severity of patients with COVID-19 with diabetes during hospitalization remains unclear. This study aimed to determine the effect of metformin on disease severity. We enrolled 110 hospitalized patients with COVID-19 with diabetes prescribed either metformin or non-metformin hypoglycemic treatment for a case-control study. The primary outcome was the occurrence of life-threatening complications. There were no differences between the two groups in age, sex, comorbidities, and clinical severity at admission. Blood glucose and lactate dehydrogenase levels of the metformin group were higher than those of the non-metformin group at admission. Other laboratory parameters at admission and treatments after admission were not different between the two groups. Strikingly, the percentage of patients who experienced life-threatening complications was significantly higher in the metformin group (28.6% (16/56) vs. 7.4% (4/54), P = 0.004). Antidiabetic therapy with metformin was associated with a higher risk of disease progression in patients with COVID-19 with diabetes during hospitalization (adjusted odds ratio = 3.964, 95% confidence interval 1.034-15.194, P = 0.045). This retrospective analysis suggested a potential safety signal for metformin, the use of which was associated with a higher risk of severe COVID-19. We propose that metformin withdrawal in patients with COVID-19 be considered to prevent disease progression.
Collapse
Affiliation(s)
- Yongchao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Tao Liu
- Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Weijun Zhong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Rong Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| |
Collapse
|
103
|
Jia H, Yue X, Lazartigues E. ACE2 mouse models: a toolbox for cardiovascular and pulmonary research. Nat Commun 2020; 11:5165. [PMID: 33057007 PMCID: PMC7560817 DOI: 10.1038/s41467-020-18880-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been identified as the host entry receptor for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) responsible for the COVID-19 pandemic. ACE2 is a regulatory enzyme of the renin-angiotensin system and has protective functions in many cardiovascular, pulmonary and metabolic diseases. This review summarizes available murine models with systemic or organ-specific deletion of ACE2, or with overexpression of murine or human ACE2. The purpose of this review is to provide researchers with the genetic tools available for further understanding of ACE2 biology and for the investigation of ACE2 in the pathogenesis and treatment of COVID-19.
Collapse
Affiliation(s)
- Hongpeng Jia
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Xinping Yue
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Eric Lazartigues
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA. .,Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA. .,Southeast Louisiana Veterans Health Care Systems, New Orleans, LA, 70119, USA.
| |
Collapse
|
104
|
Cheng X, Liu YM, Li H, Zhang X, Lei F, Qin JJ, Chen Z, Deng KQ, Lin L, Chen MM, Song X, Xia M, Huang X, Liu W, Cai J, Zhang XJ, Zhou F, Zhang P, Wang Y, Ma X, Xu Q, Yang J, Ye P, Mao W, Huang X, Xia J, Zhang BH, Guo J, Zhu L, Lu Z, Yuan Y, Wei X, She ZG, Ji YX, Li H. Metformin Is Associated with Higher Incidence of Acidosis, but Not Mortality, in Individuals with COVID-19 and Pre-existing Type 2 Diabetes. Cell Metab 2020; 32:537-547.e3. [PMID: 32861268 PMCID: PMC7439986 DOI: 10.1016/j.cmet.2020.08.013] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/23/2020] [Accepted: 08/18/2020] [Indexed: 02/08/2023]
Abstract
The safety and efficacy of anti-diabetic drugs are critical for maximizing the beneficial impacts of well-controlled blood glucose on the prognosis of individuals with COVID-19 and pre-existing type 2 diabetes (T2D). Metformin is the most commonly prescribed first-line medication for T2D, but its impact on the outcomes of individuals with COVID-19 and T2D remains to be clarified. Our current retrospective study in a cohort of 1,213 hospitalized individuals with COVID-19 and pre-existing T2D indicated that metformin use was significantly associated with a higher incidence of acidosis, particularly in cases with severe COVID-19, but not with 28-day COVID-19-related mortality. Furthermore, metformin use was significantly associated with reduced heart failure and inflammation. Our findings provide clinical evidence in support of continuing metformin treatment in individuals with COVID-19 and pre-existing T2D, but acidosis and kidney function should be carefully monitored in individuals with severe COVID-19.
Collapse
Affiliation(s)
- Xu Cheng
- Department of Cardiology, Zhongnan Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Ye-Mao Liu
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haomiao Li
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Fang Lei
- Department of Cardiology, Zhongnan Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Juan-Juan Qin
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ze Chen
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke-Qiong Deng
- Department of Cardiology, Zhongnan Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Lijin Lin
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ming-Ming Chen
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaohui Song
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Meng Xia
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Xuewei Huang
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weifang Liu
- Department of Cardiology, Zhongnan Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Xiao-Jing Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Zhou
- Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Peng Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yibin Wang
- Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19004, USA
| | - Qingbo Xu
- Centre for Clinic Pharmacology, The William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Juan Yang
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ping Ye
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiming Mao
- Department of General Surgery, Huanggang Central Hospital, Huanggang, China
| | - Xiaodong Huang
- Department of Gastroenterology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing-Hong Zhang
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine & Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China & Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Lihua Zhu
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhi-Gang She
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Yan-Xiao Ji
- Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Hongliang Li
- Department of Cardiology, Zhongnan Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
105
|
Li J, Wei Q, Li WX, McCowen KC, Xiong W, Liu J, Jiang W, Marin T, Thomas RL, He M, Gongol B, Hepokoski M, Yuan JXJ, Shyy JYJ, Xiong N, Malhotra A. Metformin Use in Diabetes Prior to Hospitalization: Effects on Mortality in Covid-19. Endocr Pract 2020; 26:1166-1172. [PMID: 33471718 PMCID: PMC7834011 DOI: 10.4158/ep-2020-0466] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 09/15/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Although type 2 diabetes mellitus (T2DM) has been reported as a risk factor for coronavirus disease 2019 (COVID-19), the effect of pharmacologic agents used to treat T2DM, such as metformin, on COVID-19 outcomes remains unclear. Metformin increases the expression of angiotensin converting enzyme 2, a known receptor for severe acute respiratory syndrome coronavirus 2. Data from people with T2DM hospitalized for COVID-19 were used to test the hypothesis that metformin use is associated with improved survival in this population. METHODS Retrospective analyses were performed on de-identified clinical data from a major hospital in Wuhan, China, that included patients with T2DM hospitalized for COVID-19 during the recent epidemic. One hundred and thirty-one patients diagnosed with COVID-19 and T2DM were used in this study. The primary outcome was mortality. Demographic, clinical characteristics, laboratory data, diabetes medications, and respiratory therapy data were also included in the analysis. RESULTS Of these 131 patients, 37 used metformin with or without other antidiabetes medications. Among the 37 metformin-taking patients, 35 (94.6%) survived and 2 (5.4%) did not survive. The mortality rates in the metformin-taking group versus the non-metformin group were 5.4% (2/37) versus 22.3% (21/94). Using multivariate analysis, metformin was found to be an independent predictor of survival in this cohort (P = .02). CONCLUSION This study reveals a significant association between metformin use and survival in people with T2DM diagnosed with COVID-19. These clinical data are consistent with potential benefits of the use of metformin for COVID-19 patients with T2DM.
Collapse
Affiliation(s)
- Jinghong Li
- From the Department of Medicine, University of California, San Diego, La Jolla, California
| | - Qi Wei
- the Department of Endocrinology, Wuhan Red Cross Hospital, Wuhan, Hubei, China
| | - Willis X Li
- From the Department of Medicine, University of California, San Diego, La Jolla, California
| | - Karen C McCowen
- From the Department of Medicine, University of California, San Diego, La Jolla, California
| | - Wei Xiong
- the Department of Endocrinology, Wuhan Red Cross Hospital, Wuhan, Hubei, China
| | - Jiao Liu
- the Department of Endocrinology, Wuhan Red Cross Hospital, Wuhan, Hubei, China
| | - Wenlijun Jiang
- the Department of Endocrinology, Wuhan Red Cross Hospital, Wuhan, Hubei, China
| | - Traci Marin
- the Department of Health Sciences, Victor Valley College, Victorville, California
| | - Robert L Thomas
- From the Department of Medicine, University of California, San Diego, La Jolla, California
| | - Ming He
- From the Department of Medicine, University of California, San Diego, La Jolla, California
| | - Brendan Gongol
- From the Department of Medicine, University of California, San Diego, La Jolla, California
| | - Mark Hepokoski
- From the Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jason X-J Yuan
- From the Department of Medicine, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- From the Department of Medicine, University of California, San Diego, La Jolla, California.
| | - Nian Xiong
- the Department of Endocrinology, Wuhan Red Cross Hospital, Wuhan, Hubei, China; the Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China..
| | - Atul Malhotra
- From the Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
106
|
Cornejo-Pareja IM, Gómez-Pérez AM, Fernández-García JC, Barahona San Millan R, Aguilera Luque A, de Hollanda A, Jiménez A, Jimenez-Murcia S, Munguia L, Ortega E, Fernandez-Aranda F, Fernández Real JM, Tinahones F. Coronavirus disease 2019 (COVID-19) and obesity. Impact of obesity and its main comorbidities in the evolution of the disease. EUROPEAN EATING DISORDERS REVIEW 2020; 28:799-815. [PMID: 32974994 DOI: 10.1002/erv.2770] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 06/22/2020] [Accepted: 06/29/2020] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic is posing a great challenge worldwide. Its rapid progression has caused thousands of deaths worldwide. Although multiple aspects remain to be clarified, some risk factors associated with a worse prognosis have been identified. These include obesity and some of its main complications, such as diabetes and high blood pressure. Furthermore, although the possible long-term complications and psychological effects that may appear in survivors of COVID-19 are not well known yet, there is a concern that those complications may be greater in obese patients. In this manuscript, we review some of the data published so far and the main points that remain to be elucidated are emphasized.
Collapse
Affiliation(s)
- Isabel M Cornejo-Pareja
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga (IBIMA), Virgen de la Victoria University Hospital, Málaga, Spain
| | - Ana M Gómez-Pérez
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga (IBIMA), Virgen de la Victoria University Hospital, Málaga, Spain
| | - José C Fernández-García
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga (IBIMA), Virgen de la Victoria University Hospital, Málaga, Spain
| | - Rebeca Barahona San Millan
- Unit of Diabetes, Endocrinology and Nutrition, Hospital de Girona Dr. Josep Trueta, 17007, Institut d'Investigació Biomèdica de Girona (IDIBGI) Girona, and Department of Medical Sciences, University of Girona, Girona, Spain
| | - Alexandre Aguilera Luque
- Unit of Diabetes, Endocrinology and Nutrition, Hospital de Girona Dr. Josep Trueta, 17007, Institut d'Investigació Biomèdica de Girona (IDIBGI) Girona, and Department of Medical Sciences, University of Girona, Girona, Spain
| | - Ana de Hollanda
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute-IDIBAPS, Hospital Clínic of Barcelona, Barcelona, Spain.,CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), ISCIII, Madrid, Spain
| | - Amanda Jiménez
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute-IDIBAPS, Hospital Clínic of Barcelona, Barcelona, Spain.,CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), ISCIII, Madrid, Spain
| | - Susana Jimenez-Murcia
- CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), ISCIII, Madrid, Spain.,Department of Psychiatry, University Hospital of Bellvitge-IDIBELL and Department of Clinical Sciences, School of Medicine and Health Sciences. University of Barcelona, Barcelona, Spain
| | - Lucero Munguia
- Department of Psychiatry, University Hospital of Bellvitge-IDIBELL and Department of Clinical Sciences, School of Medicine and Health Sciences. University of Barcelona, Barcelona, Spain
| | - Emilio Ortega
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute-IDIBAPS, Hospital Clínic of Barcelona, Barcelona, Spain.,CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), ISCIII, Madrid, Spain
| | - Fernando Fernandez-Aranda
- CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), ISCIII, Madrid, Spain.,Department of Psychiatry, University Hospital of Bellvitge-IDIBELL and Department of Clinical Sciences, School of Medicine and Health Sciences. University of Barcelona, Barcelona, Spain
| | - José M Fernández Real
- Unit of Diabetes, Endocrinology and Nutrition, Hospital de Girona Dr. Josep Trueta, 17007, Institut d'Investigació Biomèdica de Girona (IDIBGI) Girona, and Department of Medical Sciences, University of Girona, Girona, Spain.,CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), ISCIII, Madrid, Spain
| | - Francisco Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga (IBIMA), Virgen de la Victoria University Hospital, Málaga, Spain.,CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), ISCIII, Madrid, Spain
| |
Collapse
|
107
|
Wang J, Lian G, Luo L, Wang T, Xu C, Wang H, Xie L. Role of 20-hydroxyeicosatetraenoic acid in pulmonary hypertension and proliferation of pulmonary arterial smooth muscle cells. Pulm Pharmacol Ther 2020; 64:101948. [PMID: 32949704 DOI: 10.1016/j.pupt.2020.101948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 08/30/2020] [Accepted: 09/13/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To investigate the level of 20-Hydroxyeicosatetraenoic acid (20-HETE) in model of pulmonary hypertension (PH) and its effect on the proliferation of pulmonary arterial smooth muscle cells (PASMCs). METHODS Twenty male Sprague-Dawley rats were randomly divided into two groups, including control group and PH group. PH was induced by intra-peritoneal injection of 20 mg/kg monocrotaline (MCT) twice in a week in 10 rats, and control rats were given equal amount of saline. Mean pulmonary arterial pressure (mPAP), right ventricular hypertrophy index (RVHI) and pulmonary vascular remodeling index (WA%, WT%) were assessed at the week 4. The levels of 20-HETE were analysed by liquid chromatography tandem-mass spectrometry (LC-MS/MS). EdU test was used to determine the proliferation of PASMCs. Intracellular levels of reactive oxygen species (ROS) were detected using DCFH-DA dye. RESULTS (1) Prominent right ventricular hypertrophy and pulmonary vascular remodeling were verified in PH rats; (2) 20-HETE levels in lung tissue and serum were significantly lifted in PH rats; (3) Increased 20-HETE levels in cell culture supernatants were significantly noted in hypoxia condition; (4) Proliferation of PASMCs was induced by 20-HETE and hypoxia, and was inhibited by HET0016; (5) Production of ROS was elevated by 20-HETE and hypoxia, and was reduced by HET0016; CONCLUSION: Vascular remodeling was demonstrated in PH rats. 20-HETE levels were significantly increased in PH rats and under hypoxia condition. PASMCs proliferation and ROS production were elevated by 20-HETE and could be inhibited by HET0016, a specific inhibitor of 20-HETE. Taken together, changes in the level of 20-HETE may be related to the excessive proliferation of PASMCs in PH rats.
Collapse
Affiliation(s)
- Jinhua Wang
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China.
| | - Guili Lian
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China
| | - Li Luo
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China
| | - Tingjun Wang
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China
| | - Changsheng Xu
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China
| | - Huajun Wang
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China
| | - Liangdi Xie
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China.
| |
Collapse
|
108
|
Zhao Y, Zhao Z, Wang Y, Zhou Y, Ma Y, Zuo W. Single-Cell RNA Expression Profiling of ACE2, the Receptor of SARS-CoV-2. Am J Respir Crit Care Med 2020; 202:756-759. [PMID: 32663409 PMCID: PMC7462411 DOI: 10.1164/rccm.202001-0179le] [Citation(s) in RCA: 581] [Impact Index Per Article: 145.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Yu Zhao
- Tongji University Shanghai, China
| | | | | | | | - Yu Ma
- Regend Therapeutics Suzhou, China
| | - Wei Zuo
- Tongji University Shanghai, China.,The First Affiliated Hospital of Guangzhou Medical University Guangzhou, China and.,Ningxia Medical University Yinchuan, China
| |
Collapse
|
109
|
Gross LZF, Sacerdoti M, Piiper A, Zeuzem S, Leroux AE, Biondi RM. ACE2, the Receptor that Enables Infection by SARS-CoV-2: Biochemistry, Structure, Allostery and Evaluation of the Potential Development of ACE2 Modulators. ChemMedChem 2020; 15:1682-1690. [PMID: 32663362 PMCID: PMC7405163 DOI: 10.1002/cmdc.202000368] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/13/2020] [Indexed: 01/07/2023]
Abstract
Angiotensin converting enzyme 2 (ACE2) is the human receptor that interacts with the spike protein of coronaviruses, including the one that produced the 2020 coronavirus pandemic (COVID-19). Thus, ACE2 is a potential target for drugs that disrupt the interaction of human cells with SARS-CoV-2 to abolish infection. There is also interest in drugs that inhibit or activate ACE2, that is, for cardiovascular disorders or colitis. Compounds binding at alternative sites could allosterically affect the interaction with the spike protein. Herein, we review biochemical, chemical biology, and structural information on ACE2, including the recent cryoEM structures of full-length ACE2. We conclude that ACE2 is very dynamic and that allosteric drugs could be developed to target ACE2. At the time of the 2020 pandemic, we suggest that available ACE2 inhibitors or activators in advanced development should be tested for their ability to allosterically displace the interaction between ACE2 and the spike protein.
Collapse
Affiliation(s)
- Lissy Z. F. Gross
- Chemical Biology of Regulatory MechanismsIBioBA-CONICET-Partner Institute of the Max Planck SocietyGodoy Cruz 2390Buenos AiresArgentina
| | - Mariana Sacerdoti
- Chemical Biology of Regulatory MechanismsIBioBA-CONICET-Partner Institute of the Max Planck SocietyGodoy Cruz 2390Buenos AiresArgentina
| | - Albrecht Piiper
- Internal Medicine IFrankfurt University HospitalTheodor-Stern-Kai 7Frankfurt am MainGermany
| | - Stefan Zeuzem
- Internal Medicine IFrankfurt University HospitalTheodor-Stern-Kai 7Frankfurt am MainGermany
| | - Alejandro E. Leroux
- Chemical Biology of Regulatory MechanismsIBioBA-CONICET-Partner Institute of the Max Planck SocietyGodoy Cruz 2390Buenos AiresArgentina
| | - Ricardo M. Biondi
- Chemical Biology of Regulatory MechanismsIBioBA-CONICET-Partner Institute of the Max Planck SocietyGodoy Cruz 2390Buenos AiresArgentina
- Internal Medicine IFrankfurt University HospitalTheodor-Stern-Kai 7Frankfurt am MainGermany
| |
Collapse
|
110
|
Molecular mechanisms involved in the positive effects of physical activity on coping with COVID-19. Eur J Appl Physiol 2020; 120:2569-2582. [PMID: 32885275 PMCID: PMC7471545 DOI: 10.1007/s00421-020-04484-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Physical activity (PA) represents the first line of defence against diseases characterised by increased inflammation status, such as metabolic and infectious diseases. Conversely, a sedentary lifestyle-associated with obesity, type 2 diabetes and cardiovascular disorders-negatively impacts on general health status, including susceptibility to infections. At a time of a pandemic SARS-CoV2 infection, and in the context of the multiorgan crosstalk (widely accepted as a mechanism participating in the pathophysiology of all organs and systems), we examine the complex interplay mediated by skeletal muscle contraction involving the immune system and how this contributes to control health status and to counteract viral infections. In so doing, we review the molecular mechanisms and expression of molecules modulated by PA, able to provide the proper molecular equipment against viral infections such as the current SARS-CoV2. METHODS A critical review of the literature was performed to elucidate the molecular mechanisms and mediators induced by PA that potentially impact on viral infections such as SARS-CoV2. RESULTS We showed the effects mediated by regular moderate PA on viral adverse effects through the regulation of biological processes involving the crosstalk between skeletal muscle, the immune system and adipose tissue. Evidence was provided of the effects mediated by modulation of the expression of inflammation markers. CONCLUSION A tigth association between PA and reduction in inflammation status allows effective counteracting of SARS-CoV2 infection. It is therefore essential to persuade people to keep active.
Collapse
|
111
|
Zhao Y, Zhao Z, Wang Y, Zhou Y, Ma Y, Zuo W. Single-Cell RNA Expression Profiling of ACE2, the Receptor of SARS-CoV-2. Am J Respir Crit Care Med 2020. [PMID: 32663409 DOI: 10.1101/2020.01.26.919985] [Citation(s) in RCA: 265] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
AbstractA novel coronavirus SARS-CoV-2 was identified in Wuhan, Hubei Province, China in December of 2019. According to WHO report, this new coronavirus has resulted in 76,392 confirmed infections and 2,348 deaths in China by 22 February, 2020, with additional patients being identified in a rapidly growing number internationally. SARS-CoV-2 was reported to share the same receptor, Angiotensin-converting enzyme 2 (ACE2), with SARS-CoV. Here based on the public database and the state-of-the-art single-cell RNA-Seq technique, we analyzed the ACE2 RNA expression profile in the normal human lungs. The result indicates that the ACE2 virus receptor expression is concentrated in a small population of type II alveolar cells (AT2). Surprisingly, we found that this population of ACE2-expressing AT2 also highly expressed many other genes that positively regulating viral entry, reproduction and transmission. This study provides a biological background for the epidemic investigation of the COVID-19, and could be informative for future anti-ACE2 therapeutic strategy development.
Collapse
Affiliation(s)
- Yu Zhao
- Tongji University Shanghai, China
| | | | | | | | - Yu Ma
- Regend Therapeutics Suzhou, China
| | - Wei Zuo
- Tongji University Shanghai, China
- The First Affiliated Hospital of Guangzhou Medical University Guangzhou, China and
- Ningxia Medical University Yinchuan, China
| |
Collapse
|
112
|
Khan N. Possible protective role of 17β-estradiol against COVID-19. JOURNAL OF ALLERGY AND INFECTIOUS DISEASES 2020; 1:38-48. [PMID: 33196058 PMCID: PMC7665224 DOI: 10.46439/allergy.1.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) is the virus that causes coronavirus disease 2019 (COVID-19); a worldwide pandemic as declared by the World Health Organization (WHO). SARS-CoV-2 appears to infect cells by first binding and priming its viral-spike proteins with membrane-associated angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2). Through the coordinated actions of ACE2 and TMPRSS2, SARS-CoV-2 spike proteins fuse with plasma membranes and ultimately the virus enters cells. ACE2 is integral to the renin-angiotensin-aldosterone system (RAAS), and SARS-CoV-2 down-regulates protein expression levels of ACE2. Once infected, patients typically develop acute respiratory distress syndrome (ARDS) and a number of other severe complications that result in a high rate of fatality, especially in older (>60 years) adults and in people with pre-existing medical conditions. Data now indicate clearly that among people of all age groups, COVID-19 fatalities are higher in men than women. Here, attention is focused on these sex differences and posit a role of estrogen in these differences as well as possible therapeutic and protective actions of 17β-estradiol against COVID-19.
Collapse
Affiliation(s)
- Nabab Khan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| |
Collapse
|
113
|
Chen X, Guo H, Qiu L, Zhang C, Deng Q, Leng Q. Immunomodulatory and Antiviral Activity of Metformin and Its Potential Implications in Treating Coronavirus Disease 2019 and Lung Injury. Front Immunol 2020; 11:2056. [PMID: 32973814 PMCID: PMC7461864 DOI: 10.3389/fimmu.2020.02056] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/28/2020] [Indexed: 01/08/2023] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19), a disease which causes severe lung injury and multiple organ damage, presents an urgent need for new drugs. The case severity and fatality of COVID-19 are associated with excessive inflammation, namely, a cytokine storm. Metformin, a widely used drug to treat type 2 diabetes (T2D) mellitus and metabolic syndrome, has immunomodulatory activity that reduces the production of proinflammatory cytokines using macrophages and causes the formation of neutrophil extracellular traps (NETs). Metformin also inhibits the cytokine production of pathogenic Th1 and Th17 cells. Importantly, treatment with metformin alleviates various lung injuries in preclinical animal models. In addition, a recent proteomic study revealed that metformin has the potential to directly inhibit SARS-CoV-2 infection. Furthermore, retrospective clinical studies have revealed that metformin treatment reduces the mortality of T2D with COVID-19. Therefore, metformin has the potential to be repurposed to treat patients with COVID-19 at risk of developing severe illness. This review summarizes the immune pathogenesis of SARS-CoV-2 and addresses the effects of metformin on inhibiting cytokine storms and preventing SARS-CoV-2 infection, as well as its side effects.
Collapse
Affiliation(s)
- Xianyang Chen
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Huifang Guo
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Li Qiu
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Chengdong Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Qiang Deng
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qibin Leng
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
114
|
Zbinden‐Foncea H, Francaux M, Deldicque L, Hawley JA. Does High Cardiorespiratory Fitness Confer Some Protection Against Proinflammatory Responses After Infection by SARS-CoV-2? Obesity (Silver Spring) 2020; 28:1378-1381. [PMID: 32324968 PMCID: PMC7264673 DOI: 10.1002/oby.22849] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) originated in China in late 2019 and has since spread rapidly to every continent in the world. This pandemic continues to cause widespread personal suffering, along with severe pressure on medical and health care providers. The symptoms of SARS-CoV-2 and the subsequent prognosis are worsened in individuals who have preexisting comorbidities prior to infection by the virus. Individuals with obesity or overweight, insulin resistance, and diabetes typically have chronic low-grade inflammation characterized by increased levels of several proinflammatory cytokines and the inflammasome; this state predisposes to greater risk for infection along with more adverse outcomes. Here, we consider whether a high level of cardiorespiratory fitness induced by prior exercise training may confer some innate immune protection against COVID-19 by attenuating the "cytokine storm syndrome" often experienced by "at risk" individuals.
Collapse
Affiliation(s)
- Hermann Zbinden‐Foncea
- School of KinesiologyFaculty of MedicineUniversidad Finis TerraeSantiagoChile
- Centro de Salud DeportivaClinica Santa MariaSantiagoChile
- Institute of NeuroscienceUCLouvainLouvain‐la‐NeuveBelgium
| | - Marc Francaux
- Institute of NeuroscienceUCLouvainLouvain‐la‐NeuveBelgium
| | | | - John A. Hawley
- Exercise and Nutrition Research GroupMary MacKillop Institute for Health ResearchAustralian Catholic UniversityMelbourneVICAustralia
| |
Collapse
|
115
|
Jeong IK, Yoon KH, Lee MK. Diabetes and COVID-19: Global and regional perspectives. Diabetes Res Clin Pract 2020; 166:108303. [PMID: 32623038 PMCID: PMC7332438 DOI: 10.1016/j.diabres.2020.108303] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/19/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
The coronavirus disease-2019 (COVID-19) has been designated as a highly contagious infectious disease caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) since December 2019, when an outbreak of pneumonia cases emerged in Wuhan, China. The COVID-19 pandemic has led to a global health crisis, devastating the social, economic and political aspects of life. Many clinicians, health professionals, scientists, organizations, and governments have actively defeated COVID-19 and shared their experiences of the SARS-CoV2. Diabetes is one of the major risk factors for fatal outcomes from COVID-19. Patients with diabetes are vulnerable to infection because of hyperglycemia; impaired immune function; vascular complications; and comorbidities such as hypertension, dyslipidemia, and cardiovascular disease. In addition, angiotensin-converting enzyme 2 (ACE2) is a receptor for SARS-CoV-2 in the human body. Hence, the use of angiotensin-directed medications in patients with diabetes requires attention. The severity and mortality from COVID-19 was significantly higher in patients with diabetes than in those without. Thus, the patients with diabetes should take precautions during the COVID-19 pandemic. Therefore, we review the current knowledge of COVID-19 including the global and regional epidemiology, virology, impact of diabetes on COVID-19, treatment of COVID-19, and standard of care in the management of diabetes during this critical period.
Collapse
Affiliation(s)
- In-Kyung Jeong
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea.
| | - Kun Ho Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Moon Kyu Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunghyang University College of Medicine, Gumi, Republic of Korea.
| |
Collapse
|
116
|
Malhotra A, Hepokoski M, McCowen KC, Y-J Shyy J. ACE2, Metformin, and COVID-19. iScience 2020; 23:101425. [PMID: 32818905 PMCID: PMC7452173 DOI: 10.1016/j.isci.2020.101425] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/13/2020] [Accepted: 07/28/2020] [Indexed: 01/22/2023] Open
Abstract
COVID-19 is becoming a leading cause of mortality throughout the world, and few effective therapies are currently available. Angiotensin converting enzyme 2 (ACE2) is essential to COVID-19 pathogenesis, as the binding of SARS-CoV-2 spike protein (S protein) is required for viral entry and development of COVID-19. ACE2 regulates the protective arm of the renin-angiotensin-aldosterone system (RAAS) that endows anti-hypertensive and anti-inflammatory effects in the cardiovascular and pulmonary systems. Preclinical data suggest ACE2 might be downregulated after SARS-CoV-2 binding, and treatments that increase ACE2 may prevent cardiopulmonary injury. Development, testing, and mass production of novel ACE2 therapies may take years, whereas more effective treatments for COVID-19 are needed urgently. Metformin is a widely available anti-diabetic agent that has an excellent safety profile, and clinical and preclinical data suggest metformin may offer cardiopulmonary protection in COVID-19 via enhanced ACE2 expression.
Collapse
Affiliation(s)
- Atul Malhotra
- Department of Medicine, University of California, San Diego, La Jolla, CA 92023, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA 92023, USA.
| | - Mark Hepokoski
- Department of Medicine, University of California, San Diego, La Jolla, CA 92023, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA 92023, USA; VA San Diego Health System, 9500 Gilman Drive, La Jolla, CA 92023, USA
| | - Karen C McCowen
- Department of Medicine, University of California, San Diego, La Jolla, CA 92023, USA; Division of Endocrinology, University of California, San Diego, La Jolla, CA 92023, USA
| | - John Y-J Shyy
- Department of Medicine, University of California, San Diego, La Jolla, CA 92023, USA; Division of Cardiology, University of California, San Diego, La Jolla, CA 92023, USA.
| |
Collapse
|
117
|
Nuche J, Segura de la Cal T, Jiménez López Guarch C, López-Medrano F, Delgado CPO, Ynsaurriaga FA, Delgado JF, Ibáñez B, Oliver E, Subías PE. Effect of Coronavirus Disease 2019 in Pulmonary Circulation. The Particular Scenario of Precapillary Pulmonary Hypertension. Diagnostics (Basel) 2020; 10:E548. [PMID: 32752129 PMCID: PMC7459745 DOI: 10.3390/diagnostics10080548] [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: 07/20/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023] Open
Abstract
The Coronavirus Disease of 2019 (COVID-19) has supposed a global health emergency affecting millions of people, with particular severity in the elderly and patients with previous comorbidities, especially those with cardiovascular disease. Patients with pulmonary arterial hypertension (PAH) and chronic thromboembolic pulmonary hypertension (CTEPH) could represent an especially vulnerable population because of the high mortality rates reported for respiratory infections. However, the number of COVID-19 cases reported among PAH and CTEPH patients is surprisingly low. Furthermore, the clinical picture that has been described in these patients is far from the severity that experts would expect. Endothelial dysfunction is a common feature between patients with PAH/CTEPH and COVID-19, leading to ventilation/perfusion mismatch, vasoconstriction, thrombosis and inflammation. In this picture, the angiotensin-converting enzyme 2 plays an essential role, being directly involved in the pathophysiology of both clinical entities. Some of these common characteristics could explain the good adaptation of PAH and CTEPH patients to COVID-19, who could also have obtained a benefit from the disease's specific treatments (anticoagulant and pulmonary vasodilators), probably due to its protective effect on the endothelium. Additionally, these common features could also lead to PAH/CTEPH as a potential sequelae of COVID-19. Throughout this comprehensive review, we describe the similarities and differences between both conditions and the possible pathophysiological and therapeutic-based mechanisms leading to the low incidence and severity of COVID-19 reported in PAH/CTEPH patients to date. Nevertheless, international registries should look carefully into this population for better understanding and management.
Collapse
Affiliation(s)
- Jorge Nuche
- Centro de Investigaciones Biomédicas En Red de enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.N.); (C.J.L.G.); (F.A.Y.); (J.F.D.); (B.I.)
- Servicio de Cardiología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), 28041 Madrid, Spain; (T.S.d.l.C.); (C.P.-O.D.)
- Centro Nacional de Investigaciones Cardiovasculares, 28029 Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Teresa Segura de la Cal
- Servicio de Cardiología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), 28041 Madrid, Spain; (T.S.d.l.C.); (C.P.-O.D.)
| | - Carmen Jiménez López Guarch
- Centro de Investigaciones Biomédicas En Red de enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.N.); (C.J.L.G.); (F.A.Y.); (J.F.D.); (B.I.)
- Servicio de Cardiología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), 28041 Madrid, Spain; (T.S.d.l.C.); (C.P.-O.D.)
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Francisco López-Medrano
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
- Department of Infectious Diseases, Hospital Universitario 12 de Octubre, Instituto de Investigacioón Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Carmen Pérez-Olivares Delgado
- Servicio de Cardiología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), 28041 Madrid, Spain; (T.S.d.l.C.); (C.P.-O.D.)
| | - Fernando Arribas Ynsaurriaga
- Centro de Investigaciones Biomédicas En Red de enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.N.); (C.J.L.G.); (F.A.Y.); (J.F.D.); (B.I.)
- Servicio de Cardiología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), 28041 Madrid, Spain; (T.S.d.l.C.); (C.P.-O.D.)
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Juan F. Delgado
- Centro de Investigaciones Biomédicas En Red de enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.N.); (C.J.L.G.); (F.A.Y.); (J.F.D.); (B.I.)
- Servicio de Cardiología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), 28041 Madrid, Spain; (T.S.d.l.C.); (C.P.-O.D.)
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Borja Ibáñez
- Centro de Investigaciones Biomédicas En Red de enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.N.); (C.J.L.G.); (F.A.Y.); (J.F.D.); (B.I.)
- Centro Nacional de Investigaciones Cardiovasculares, 28029 Madrid, Spain
- IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Eduardo Oliver
- Centro de Investigaciones Biomédicas En Red de enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.N.); (C.J.L.G.); (F.A.Y.); (J.F.D.); (B.I.)
- Centro Nacional de Investigaciones Cardiovasculares, 28029 Madrid, Spain
| | - Pilar Escribano Subías
- Centro de Investigaciones Biomédicas En Red de enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.N.); (C.J.L.G.); (F.A.Y.); (J.F.D.); (B.I.)
- Servicio de Cardiología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), 28041 Madrid, Spain; (T.S.d.l.C.); (C.P.-O.D.)
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| |
Collapse
|
118
|
Shen H, Zhang J, Wang C, Jain PP, Xiong M, Shi X, Lei Y, Chen S, Yin Q, Thistlethwaite PA, Wang J, Gong K, Yuan ZY, Yuan JXJ, Shyy JYJ. MDM2-Mediated Ubiquitination of Angiotensin-Converting Enzyme 2 Contributes to the Development of Pulmonary Arterial Hypertension. Circulation 2020; 142:1190-1204. [PMID: 32755395 PMCID: PMC7497891 DOI: 10.1161/circulationaha.120.048191] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Angiotensin-converting enzyme 2 (ACE2) converts angiotensin II, a potent vasoconstrictor, to angiotensin-(1–7) and is also a membrane protein that enables coronavirus disease 2019 (COVID-19) infectivity. AMP-activated protein kinase (AMPK) phosphorylation of ACE2 enhances ACE2 stability. This mode of posttranslational modification of ACE2 in vascular endothelial cells is causative of a pulmonary hypertension (PH)–protective phenotype. The oncoprotein MDM2 (murine double minute 2) is an E3 ligase that ubiquitinates its substrates to cause their degradation. In this study, we investigated whether MDM2 is involved in the posttranslational modification of ACE2 through its ubiquitination of ACE2, and whether an AMPK and MDM2 crosstalk regulates the pathogenesis of PH. Methods: Bioinformatic analyses were used to explore E3 ligase that ubiquitinates ACE2. Cultured endothelial cells, mouse models, and specimens from patients with idiopathic pulmonary arterial hypertension were used to investigate the crosstalk between AMPK and MDM2 in regulating ACE2 phosphorylation and ubiquitination in the context of PH. Results: Levels of MDM2 were increased and those of ACE2 decreased in lung tissues or pulmonary arterial endothelial cells from patients with idiopathic pulmonary arterial hypertension and rodent models of experimental PH. MDM2 inhibition by JNJ-165 reversed the SU5416/hypoxia-induced PH in C57BL/6 mice. ACE2-S680L mice (dephosphorylation at S680) showed PH susceptibility, and ectopic expression of ACE2-S680L/K788R (deubiquitination at K788) reduced experimental PH. Moreover, ACE2-K788R overexpression in mice with endothelial cell–specific AMPKα2 knockout mitigated PH. Conclusions: Maladapted posttranslational modification (phosphorylation and ubiquitination) of ACE2 at Ser-680 and Lys-788 is involved in the pathogenesis of pulmonary arterial hypertension and experimental PH. Thus, a combined intervention of AMPK and MDM2 in the pulmonary endothelium might be therapeutically effective in PH treatment.
Collapse
Affiliation(s)
- Hui Shen
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, China (H.S., K.G.)
| | - Jiao Zhang
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (J.Z., C.W., Y.L., Z.-Y.Y.).,Division of Cardiology, Department of Medicine (J.Z., J.Y.-J.S.), University of California, San Diego, La Jolla.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (J.Z., C.W., Y.L., S.C., Q.Y.)
| | - Chen Wang
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (J.Z., C.W., Y.L., Z.-Y.Y.).,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (J.Z., C.W., Y.L., S.C., Q.Y.)
| | - Pritesh P Jain
- Division of Pulmonary, Critical Care and Sleep Medicine (P.P.J., M.X., J.W., J.X.-J.Y.), University of California, San Diego, La Jolla
| | - Mingmei Xiong
- Division of Pulmonary, Critical Care and Sleep Medicine (P.P.J., M.X., J.W., J.X.-J.Y.), University of California, San Diego, La Jolla.,Department of Critical Medicine, The Third Affiliated Hospital of Guangzhou Medical University, China (M.X.)
| | | | - Yuyang Lei
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (J.Z., C.W., Y.L., Z.-Y.Y.).,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (J.Z., C.W., Y.L., S.C., Q.Y.)
| | - Shanshan Chen
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (J.Z., C.W., Y.L., S.C., Q.Y.)
| | - Qian Yin
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (J.Z., C.W., Y.L., S.C., Q.Y.)
| | - Patricia A Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery (P.A.T.), University of California, San Diego, La Jolla
| | - Jian Wang
- Division of Pulmonary, Critical Care and Sleep Medicine (P.P.J., M.X., J.W., J.X.-J.Y.), University of California, San Diego, La Jolla.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, China (J.W.)
| | - Kaizheng Gong
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, China (H.S., K.G.)
| | - Zu-Yi Yuan
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (J.Z., C.W., Y.L., Z.-Y.Y.)
| | - Jason X-J Yuan
- Division of Pulmonary, Critical Care and Sleep Medicine (P.P.J., M.X., J.W., J.X.-J.Y.), University of California, San Diego, La Jolla
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine (J.Z., J.Y.-J.S.), University of California, San Diego, La Jolla
| |
Collapse
|
119
|
Amraei R, Rahimi N. COVID-19, Renin-Angiotensin System and Endothelial Dysfunction. Cells 2020; 9:E1652. [PMID: 32660065 PMCID: PMC7407648 DOI: 10.3390/cells9071652] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 01/08/2023] Open
Abstract
The newly emergent novel coronavirus disease 2019 (COVID-19) outbreak, which is caused by SARS-CoV-2 virus, has posed a serious threat to global public health and caused worldwide social and economic breakdown. Angiotensin-converting enzyme 2 (ACE2) is expressed in human vascular endothelium, respiratory epithelium, and other cell types, and is thought to be a primary mechanism of SARS-CoV-2 entry and infection. In physiological condition, ACE2 via its carboxypeptidase activity generates angiotensin fragments (Ang 1-9 and Ang 1-7), and plays an essential role in the renin-angiotensin system (RAS), which is a critical regulator of cardiovascular homeostasis. SARS-CoV-2 via its surface spike glycoprotein interacts with ACE2 and invades the host cells. Once inside the host cells, SARS-CoV-2 induces acute respiratory distress syndrome (ARDS), stimulates immune response (i.e., cytokine storm) and vascular damage. SARS-CoV-2 induced endothelial cell injury could exacerbate endothelial dysfunction, which is a hallmark of aging, hypertension, and obesity, leading to further complications. The pathophysiology of endothelial dysfunction and injury offers insights into COVID-19 associated mortality. Here we reviewed the molecular basis of SARS-CoV-2 infection, the roles of ACE2, RAS signaling, and a possible link between the pre-existing endothelial dysfunction and SARS-CoV-2 induced endothelial injury in COVID-19 associated mortality. We also surveyed the roles of cell adhesion molecules (CAMs), including CD209L/L-SIGN and CD209/DC-SIGN in SARS-CoV-2 infection and other related viruses. Understanding the molecular mechanisms of infection, the vascular damage caused by SARS-CoV-2 and pathways involved in the regulation of endothelial dysfunction could lead to new therapeutic strategies against COVID-19.
Collapse
Affiliation(s)
- Razie Amraei
- Department of Pathology, School of Medicine, Boston University Medical Campus, Boston, MA 02118, USA
| | - Nader Rahimi
- Department of Pathology, School of Medicine, Boston University Medical Campus, Boston, MA 02118, USA
| |
Collapse
|
120
|
Li Y, Zhou W, Yang L, You R. Physiological and pathological regulation of ACE2, the SARS-CoV-2 receptor. Pharmacol Res 2020; 157:104833. [PMID: 32302706 PMCID: PMC7194807 DOI: 10.1016/j.phrs.2020.104833] [Citation(s) in RCA: 239] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023]
Abstract
The renin-angiotensin system (RAS) is crucial for the physiology and pathology of all the organs. Angiotensin-converting enzyme 2 (ACE2) maintains the homeostasis of RAS as a negative regulator. Recently, ACE2 was identified as the receptor of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the coronavirus that is causing the pandemic of Coronavirus disease 2019 (COVID-19). Since SARS-CoV-2 must bind with ACE2 before entering the host cells in humans, the distribution and expression of ACE2 may be critical for the target organ of the SARS-CoV-2 infection. Moreover, accumulating evidence has demonstrated the implication of ACE2 in the pathological progression in tissue injury and several chronic diseases, ACE2 may also be essential in the progression and clinical outcomes of COVID-19. Therefore, we summarized the expression and activity of ACE2 in various physiological and pathological conditions, and discussed its potential implication in the susceptibility of SARS-CoV-2 infection and the progression and prognosis of COVID-19 patients in the current review.
Collapse
Affiliation(s)
- Yanwei Li
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210000, China; School of Medicine, Southeast University, Nanjing, 210000, China
| | - Wei Zhou
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Li Yang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210000, China.
| | - Ran You
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210000, China.
| |
Collapse
|
121
|
Ceriello A, Standl E, Catrinoiu D, Itzhak B, Lalic NM, Rahelic D, Schnell O, Škrha J, Valensi P. Issues of Cardiovascular Risk Management in People With Diabetes in the COVID-19 Era. Diabetes Care 2020; 43:1427-1432. [PMID: 32409501 DOI: 10.2337/dc20-0941] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 02/03/2023]
Abstract
People with diabetes compared with people without exhibit worse prognosis if affected by coronavirus disease 2019 (COVID-19) induced by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), particularly when compromising metabolic control and concomitant cardiovascular disorders are present. This Perspective seeks to explore newly occurring cardio-renal-pulmonary organ damage induced or aggravated by the disease process of COVID-19 and its implications for the cardiovascular risk management of people with diabetes, especially taking into account potential interactions with mechanisms of cellular intrusion of SARS-CoV-2. Severe infection with SARS-CoV-2 can precipitate myocardial infarction, myocarditis, heart failure, and arrhythmias as well as an acute respiratory distress syndrome and renal failure. They may evolve along with multiorgan failure directly due to SARS-CoV-2-infected endothelial cells and resulting endotheliitis. This complex pathology may bear challenges for the use of most diabetes medications in terms of emerging contraindications that need close monitoring of all people with diabetes diagnosed with SARS-CoV-2 infection. Whenever possible, continuous glucose monitoring should be implemented to ensure stable metabolic compensation. Patients in the intensive care unit requiring therapy for glycemic control should be handled solely by intravenous insulin using exact dosing with a perfusion device. Although not only ACE inhibitors and angiotensin 2 receptor blockers but also SGLT2 inhibitors, GLP-1 receptor agonists, pioglitazone, and probably insulin seem to increase the number of ACE2 receptors on the cells utilized by SARS-CoV-2 for penetration, no evidence presently exists that shows this might be harmful in terms of acquiring or worsening COVID-19. In conclusion, COVID-19 and related cardio-renal-pulmonary damage can profoundly affect cardiovascular risk management of people with diabetes.
Collapse
Affiliation(s)
| | - Eberhard Standl
- Forschergruppe Diabetes e.V. at Munich Helmholtz Centre, Munich, Germany
| | - Doina Catrinoiu
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| | - Baruch Itzhak
- Clalit Health Services and Technion Faculty of Medicine, Haifa, Israel
| | - Nebojsa M Lalic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dario Rahelic
- Vuk Vrhovac University Clinic for Diabetes, Endocrinology and Metabolic Diseases, Merkur University Hospital, Zagreb, Croatia.,University of Zagreb School of Medicine, Zagreb, Croatia.,University of Osijek School of Medicine, Osijek, Croatia
| | - Oliver Schnell
- Forschergruppe Diabetes e.V. at Munich Helmholtz Centre, Munich, Germany
| | - Jan Škrha
- Department of Internal Medicine 3, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | | |
Collapse
|
122
|
Sharma RK, Oliveira AC, Yang T, Karas MM, Li J, Lobaton GO, Aquino VP, Robles-Vera I, de Kloet AD, Krause EG, Bryant AJ, Verma A, Li Q, Richards EM, Raizada MK. Gut Pathology and Its Rescue by ACE2 (Angiotensin-Converting Enzyme 2) in Hypoxia-Induced Pulmonary Hypertension. Hypertension 2020; 76:206-216. [PMID: 32418496 PMCID: PMC7505091 DOI: 10.1161/hypertensionaha.120.14931] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023]
Abstract
Therapeutic advances for pulmonary hypertension (PH) have been incremental because of the focus on the pulmonary vasculature in PH pathology. Here, we evaluate the concept that PH is, rather, a systemic disorder involving interplay among multiorgan systems, including brain, gut, and lungs. Therefore, the objective of this study was to evaluate the hypothesis that PH is associated with a dysfunctional brain-gut-lung axis and that global overexpression of ACE2 (angiotensin-converting enzyme 2) rebalances this axis and protects against PH. ACE2 knockin and wild-type (WT; C57BL/6) mice were subjected to chronic hypoxia (10% FIO2) or room air for 4 weeks. Cardiopulmonary hemodynamics, histology, immunohistochemistry, and fecal 16S rRNA microbial gene analyses were evaluated. Hypoxia significantly increased right ventricular systolic pressure, sympathetic activity as well as the number and activation of microglia in the paraventricular nucleus of the hypothalamus in WT mice. This was associated with a significant increase in muscularis layer thickening and decreases in both villi length and goblet cells and altered gut microbiota. Global overexpression of ACE2 prevented changes in hypoxia-induced pulmonary and gut pathophysiology and established distinct microbial communities from WT hypoxia mice. Furthermore, WT mice subjected to fecal matter transfer from ACE2 knockin mice were resistant to hypoxia-induced PH compared with their controls receiving WT fecal matter transfer. These observations demonstrate that ACE2 ameliorates these hypoxia-induced pathologies and attenuates PH. The data implicate dysfunctional brain-gut-lung communication in PH and provide novel avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Ravindra K. Sharma
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Aline C. Oliveira
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Tao Yang
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Marianthi M. Karas
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Jing Li
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Gilberto O. Lobaton
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Victor P. Aquino
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Iñaki Robles-Vera
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain
| | - Annette D. de Kloet
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Eric G. Krause
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Andrew J. Bryant
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Amrisha Verma
- Department of Ophthalmology Research, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Qiuhong Li
- Department of Ophthalmology Research, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Elaine M. Richards
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Mohan K. Raizada
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
123
|
Park JF, Banerjee S, Umar S. In the eye of the storm: the right ventricle in COVID-19. Pulm Circ 2020; 10:2045894020936660. [PMID: 32655856 PMCID: PMC7333504 DOI: 10.1177/2045894020936660] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023] Open
Abstract
The corona virus disease of 2019 pandemic caused by the SARS-CoV-2 virus continues to inflict significant morbidity and mortality around the globe. A variety of cardiovascular presentations of SARS-CoV-2 infection have been described so far. However, the impact of SARS-CoV-2 on the right ventricle is largely unknown. Due to its pathophysiologic relevance, the right ventricle finds itself in the eye of the storm of corona virus disease of 2019, placing it at higher risk of failure. Increased afterload from acute respiratory distress syndrome and pulmonary embolism, negative inotropic effects of cytokines, and direct angiotensin converting enzyme 2-mediated cardiac injury from SARS-CoV-2 are potential mechanisms of right ventricle dysfunction in corona virus disease of 2019. Early detection and treatment of right ventricle dysfunction may lead to decreased mortality and improved patient outcomes in corona virus disease of 2019.
Collapse
Affiliation(s)
- John F. Park
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Somanshu Banerjee
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Soban Umar
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
124
|
Shaikh SB, Prabhakar Bhandary Y. Effect of curcumin on IL-17A mediated pulmonary AMPK kinase/cyclooxygenase-2 expressions via activation of NFκB in bleomycin-induced acute lung injury in vivo. Int Immunopharmacol 2020; 85:106676. [PMID: 32535538 DOI: 10.1016/j.intimp.2020.106676] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/20/2020] [Accepted: 06/03/2020] [Indexed: 11/25/2022]
Abstract
Acute lung injury (ALI) remains to be the major cause of mortality. Bleomycin (BLM) injury activates the pro-inflammatory cytokine Interleukin L-17A which regulates the expression of COX-2 and inhibits P-AMPKα in BLM/IL-17A exposed mice upon activation of NFκB and other inflammatory molecules the actual mechanism behind which remains unclear. The current investigation was carried out to assess the role of IL-17A with COX-2 and P- AMPKα and to highlight the important contribution of adjunctive use of curcumin as a promising preventive strategy for the BLM-induced ALI. Immunofluorescence analysis reveals that the natural spice curcumin blocks the expressions of COX-2, NF-κB-p65, fibronectin (FBN), and expresses P-AMPKα in vivo. Curcumin could also suppress the expressions of NF-κB-p105 in BLM/IL-17A exposed mice. mRNA expressions showed reduced expressions of PDGFA, PDGFB, CTGF, IGF1, NFκB1, NFκB2, MMP-3, MMP-9, and MMP-14 on curcumin treatment. Our study implicates a critical role of AMPKα/COX- 2 in the emergence of pulmonary fibrosis via exerting the potential role of curcumin as an adjuvant anti-inflammatory therapeutic for treating lung injury.
Collapse
Affiliation(s)
- Sadiya Bi Shaikh
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore 575018, Karnataka, India
| | | |
Collapse
|
125
|
Ursini F, Ciaffi J, Landini MP, Meliconi R. COVID-19 and diabetes: Is metformin a friend or foe? Diabetes Res Clin Pract 2020; 164:108167. [PMID: 32339534 PMCID: PMC7195096 DOI: 10.1016/j.diabres.2020.108167] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Francesco Ursini
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum - University of Bologna, Via Massarenti, 9, 40138 Bologna, Italy; Medicine & Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, via Giulio Cesare Pupilli, 1, 40136 Bologna, Italy.
| | - Jacopo Ciaffi
- Medicine & Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, via Giulio Cesare Pupilli, 1, 40136 Bologna, Italy
| | - Maria Paola Landini
- Scientific Direction, IRCCS Istituto Ortopedico Rizzoli, via Giulio Cesare Pupilli, 1, 40136 Bologna, Italy
| | - Riccardo Meliconi
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum - University of Bologna, Via Massarenti, 9, 40138 Bologna, Italy; Medicine & Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, via Giulio Cesare Pupilli, 1, 40136 Bologna, Italy
| |
Collapse
|
126
|
Devaux CA, Rolain JM, Raoult D. ACE2 receptor polymorphism: Susceptibility to SARS-CoV-2, hypertension, multi-organ failure, and COVID-19 disease outcome. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2020; 53:425-435. [PMID: 32414646 PMCID: PMC7201239 DOI: 10.1016/j.jmii.2020.04.015] [Citation(s) in RCA: 333] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has emerged in Chinese people in December 2019 and has currently spread worldwide causing the COVID-19 pandemic with more than 150,000 deaths. In order for a SARS-CoV like virus circulating in wild life for a very long time to infect the index case-patient, a number of conditions must be met, foremost among which is the encounter with humans and the presence in homo sapiens of a cellular receptor allowing the virus to bind. Recently it was shown that the SARS-CoV-2 spike protein, binds to the human angiotensin I converting enzyme 2 (ACE2). This molecule is a peptidase expressed at the surface of lung epithelial cells and other tissues, that regulates the renin-angiotensin-aldosterone system. Humans are not equal with respect to the expression levels of the cellular ACE2. Moreover, ACE2 polymorphisms were recently described in human populations. Here we review the most recent evidence that ACE2 expression and/or polymorphism could influence both the susceptibility of people to SARS-CoV-2 infection and the outcome of the COVID-19 disease. Further exploration of the relationship between the virus, the peptidase function of ACE2 and the levels of angiotensin II in SARS-CoV-2 infected patients should help to better understand the pathophysiology of the disease and the multi-organ failures observed in severe COVID-19 cases, particularly heart failure.
Collapse
Affiliation(s)
- Christian A Devaux
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France; CNRS, Marseille, France; IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France.
| | - Jean-Marc Rolain
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France; IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | - Didier Raoult
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France; IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| |
Collapse
|
127
|
Evans AM, Hardie DG. AMPK and the Need to Breathe and Feed: What's the Matter with Oxygen? Int J Mol Sci 2020; 21:ijms21103518. [PMID: 32429235 PMCID: PMC7279029 DOI: 10.3390/ijms21103518] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
We live and to do so we must breathe and eat, so are we a combination of what we eat and breathe? Here, we will consider this question, and the role in this respect of the AMP-activated protein kinase (AMPK). Emerging evidence suggests that AMPK facilitates central and peripheral reflexes that coordinate breathing and oxygen supply, and contributes to the central regulation of feeding and food choice. We propose, therefore, that oxygen supply to the body is aligned with not only the quantity we eat, but also nutrient-based diet selection, and that the cell-specific expression pattern of AMPK subunit isoforms is critical to appropriate system alignment in this respect. Currently available information on how oxygen supply may be aligned with feeding and food choice, or vice versa, through our motivation to breathe and select particular nutrients is sparse, fragmented and lacks any integrated understanding. By addressing this, we aim to provide the foundations for a clinical perspective that reveals untapped potential, by highlighting how aberrant cell-specific changes in the expression of AMPK subunit isoforms could give rise, in part, to known associations between metabolic disease, such as obesity and type 2 diabetes, sleep-disordered breathing, pulmonary hypertension and acute respiratory distress syndrome.
Collapse
Affiliation(s)
- A. Mark Evans
- Centre for Discovery Brain Sciences and Cardiovascular Science, Edinburgh Medical School, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
- Correspondence:
| | - D. Grahame Hardie
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK;
| |
Collapse
|
128
|
Zhang J, He Y, Yan X, Chen S, He M, Lei Y, Zhang J, Gongol B, Gu M, Miao Y, Bai L, Cui X, Wang X, Zhang Y, Fan F, Li Z, Shen Y, Chou C, Huang H, Malhotra A, Rabinovitch M, Jing Z, Shyy JY. MicroRNA-483 amelioration of experimental pulmonary hypertension. EMBO Mol Med 2020; 12:e11303. [PMID: 32324970 PMCID: PMC7207157 DOI: 10.15252/emmm.201911303] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/30/2022] Open
Abstract
Endothelial dysfunction is critically involved in the pathogenesis of pulmonary arterial hypertension (PAH) and that exogenously administered microRNA may be of therapeutic benefit. Lower levels of miR-483 were found in serum from patients with idiopathic pulmonary arterial hypertension (IPAH), particularly those with more severe disease. RNA-seq and bioinformatics analyses showed that miR-483 targets several PAH-related genes, including transforming growth factor-β (TGF-β), TGF-β receptor 2 (TGFBR2), β-catenin, connective tissue growth factor (CTGF), interleukin-1β (IL-1β), and endothelin-1 (ET-1). Overexpression of miR-483 in ECs inhibited inflammatory and fibrogenic responses, revealed by the decreased expression of TGF-β, TGFBR2, β-catenin, CTGF, IL-1β, and ET-1. In contrast, inhibition of miR-483 increased these genes in ECs. Rats with EC-specific miR-483 overexpression exhibited ameliorated pulmonary hypertension (PH) and reduced right ventricular hypertrophy on challenge with monocrotaline (MCT) or Sugen + hypoxia. A reversal effect was observed in rats that received MCT with inhaled lentivirus overexpressing miR-483. These results indicate that PAH is associated with a reduced level of miR-483 and that miR-483 might reduce experimental PH by inhibition of multiple adverse responses.
Collapse
Affiliation(s)
- Jin Zhang
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterKey Laboratory of Environment and Genes Related to DiseasesMinistry of Education of ChinaXi'an Jiaotong UniversityXianChina
| | - Yangyang He
- State Key Laboratory of Cardiovascular disease & FuWai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Xiaosong Yan
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterKey Laboratory of Environment and Genes Related to DiseasesMinistry of Education of ChinaXi'an Jiaotong UniversityXianChina
| | - Shanshan Chen
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterKey Laboratory of Environment and Genes Related to DiseasesMinistry of Education of ChinaXi'an Jiaotong UniversityXianChina
| | - Ming He
- Department of MedicineUniversity of CaliforniaSan DiegoLa JollaCAUSA
| | - Yuyang Lei
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterKey Laboratory of Environment and Genes Related to DiseasesMinistry of Education of ChinaXi'an Jiaotong UniversityXianChina
| | - Jiao Zhang
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterKey Laboratory of Environment and Genes Related to DiseasesMinistry of Education of ChinaXi'an Jiaotong UniversityXianChina
- Department of MedicineUniversity of CaliforniaSan DiegoLa JollaCAUSA
- Department of CardiologyFirst Affiliated HospitalXi'an Jiaotong UniversityXianChina
| | - Brendan Gongol
- Department of MedicineUniversity of CaliforniaSan DiegoLa JollaCAUSA
| | - Mingxia Gu
- Department of Pediatrics (Cardiology)Cardiovascular Institute and Wall Center for Pulmonary Vascular DiseasesStanford University School of MedicineStanfordCAUSA
| | - Yifei Miao
- Department of Pediatrics (Cardiology)Cardiovascular Institute and Wall Center for Pulmonary Vascular DiseasesStanford University School of MedicineStanfordCAUSA
| | - Liang Bai
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterKey Laboratory of Environment and Genes Related to DiseasesMinistry of Education of ChinaXi'an Jiaotong UniversityXianChina
| | - Xiaopei Cui
- Department of Geriatric MedicineQilu Hospital of Shandong UniversityJinanChina
| | - Xiaojian Wang
- State Key Laboratory of Cardiovascular disease & FuWai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Yixin Zhang
- State Key Laboratory of Cardiovascular disease & FuWai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Fenling Fan
- Department of CardiologyFirst Affiliated HospitalXi'an Jiaotong UniversityXianChina
| | - Zhao Li
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterKey Laboratory of Environment and Genes Related to DiseasesMinistry of Education of ChinaXi'an Jiaotong UniversityXianChina
| | - Yuan Shen
- Department of Epidemiology and Health StatisticsSchool of Public HealthXi'an Jiaotong UniversityXianChina
| | - Chih‐Hung Chou
- Department of Biological Science and TechnologyNational Chiao Tung UniversityHsinchuTaiwan
| | - Hsien‐Da Huang
- Warshel Institute for Computational BiologySchool of Life and Health SciencesThe Chinese University of Hong KongShenzhenChina
| | - Atul Malhotra
- Department of MedicineUniversity of CaliforniaSan DiegoLa JollaCAUSA
| | - Marlene Rabinovitch
- Department of Pediatrics (Cardiology)Cardiovascular Institute and Wall Center for Pulmonary Vascular DiseasesStanford University School of MedicineStanfordCAUSA
| | - Zhi‐Cheng Jing
- Department of Cardiology & Key Lab of Pulmonary Vascular MedicinePeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - John Y‐J Shyy
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterKey Laboratory of Environment and Genes Related to DiseasesMinistry of Education of ChinaXi'an Jiaotong UniversityXianChina
- Department of MedicineUniversity of CaliforniaSan DiegoLa JollaCAUSA
| |
Collapse
|
129
|
Zhou H, Liu LP, Fang M, Li YM, Zheng YW. A potential ex vivo infection model of human induced pluripotent stem cell-3D organoids beyond coronavirus disease 2019. Histol Histopathol 2020; 35:1077-1082. [PMID: 32339250 DOI: 10.14670/hh-18-223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The novel coronavirus disease 2019 (COVID-19) outbreak began in the city of Wuhan, whereupon it rapidly spread throughout China and subsequently across the world. Rapid transmission of COVID-19 has caused wide-spread panic. Many established medications have been used to treat the disease symptoms; however, no specific drugs or vaccines have been developed. Organoids derived from human induced pluripotent stem cells (iPSCs) may serve as suitable infection models for ex vivo mimicking of the viral life cycle and drug screening. Human iPSC-3D organoids, self-organised tissues with multiple cell environments, have a similar structure and function as real human organs; hence, these organoids allow greater viral infection efficiency, mimic the natural host-virus interactions, and are suitable for long-term experimentation. Here, we suggest the use of a functional human iPSC-organoid that could act as a reliable and feasible ex vivo infection model for investigation of the virus. This approach will provide much needed insight into the underlying molecular dynamics of COVID-19 for the development of novel treatment and prevention strategies.
Collapse
Affiliation(s)
- Hang Zhou
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Li-Ping Liu
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mei Fang
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yu-Mei Li
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Yun-Wen Zheng
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China.,Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan. .,School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan.,Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,School of Biotechnology and Healt Sciences, Wuyi University, Jiangmen, Guandong, P.R. China
| |
Collapse
|
130
|
Liu R, Yang L, Wei Q. miR-34a targets PAI-1 to regulate urinary microalbumin and renal function in hypertensive mice. Eur J Med Res 2020; 25:3. [PMID: 32178735 PMCID: PMC7077132 DOI: 10.1186/s40001-020-00404-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 02/17/2020] [Indexed: 12/11/2022] Open
Abstract
Background The aim of the study is to investigate the effects of miR-34a targeted at PAI-1 on urinary microalbumin and renal function in hypertensive mice. Methods Twenty specific-pathogen-free (SPF) BPN/3J mice were selected in normal group, and 120 SPF BPH/2J mice were evenly divided into model group, negative control group, miR-34a mimic group, miR-34a inhibitor group, Si-PAI-1 group, and miR-34a inhibitor + Si-PAI-1 group. qRT-PCR was used to detect the expression of miR-34a and PAI-1 mRNA. The protein expressions of PAI-1, angiotensin-converting enzyme (ACE) and ACE2 were detected by Western blot. Serum levels of AngII and Ang1-7 were detected by ELISA. Results miR-34a negatively regulated the expression of PAI-1. Compared with the normal group, mice in the other groups had significantly lower body weight, increased systolic blood pressure and 24-h urinary microalbumin content, decreased miR-34a expression, superoxide dismutase (SOD) and nitric oxide (NO) content, and ACE2 protein expression, and increased PAI-1 expression, serum creatinine (Scr), blood urea nitrogen (BUN) malondialdehyde (MDA), AngII and Ang1-7 levels, and ACE protein expression (all P < 0.05). Compared with the model group, mice in the miR-34a mimic group and Si-PAI-1 group had no significant changes in body weight (all P > 0.05), while they had significantly lower systolic blood pressure and 24-h urinary microalbumin content, increased SOD and NO levels and ACE2 protein expression, and decreased PAI-1 expression, Scr, BUN, MDA, AngII and Ang1-7 levels, and ACE protein expression (all P < 0.05). Compared with the miR-34a inhibitor group, symptoms in miR-34a inhibitor + Si-PAI-1 group were significantly improved (all P < 0.05). Conclusions miR-34a can inhibit the expression of PAI-1, thereby reducing urinary microalbumin content in hypertensive mice and protecting their renal function.
Collapse
Affiliation(s)
- Ruitao Liu
- Department of Cardiovascular Medicine, Xingtai People's Hospital, No.16 Hongxing East Street, Qiaodong District, Xingtai, 054000, Hebei, China
| | - Lihong Yang
- Department of Cardiovascular Medicine, Xingtai People's Hospital, No.16 Hongxing East Street, Qiaodong District, Xingtai, 054000, Hebei, China
| | - Qingmin Wei
- Department of Cardiovascular Medicine, Xingtai People's Hospital, No.16 Hongxing East Street, Qiaodong District, Xingtai, 054000, Hebei, China.
| |
Collapse
|
131
|
Zahid KR, Raza U, Chen J, Raj UJ, Gou D. Pathobiology of pulmonary artery hypertension: role of long non-coding RNAs. Cardiovasc Res 2020; 116:1937-1947. [PMID: 32109276 DOI: 10.1093/cvr/cvaa050] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/30/2019] [Accepted: 02/25/2020] [Indexed: 12/30/2022] Open
Abstract
Abstract
Pulmonary arterial hypertension (PAH) is a disease with complex pathobiology, significant morbidity and mortality, and remains without a cure. It is characterized by vascular remodelling associated with uncontrolled proliferation of pulmonary artery smooth muscle cells, endothelial cell proliferation and dysfunction, and endothelial-to-mesenchymal transition, leading to narrowing of the vascular lumen, increased vascular resistance and pulmonary arterial pressure, which inevitably results in right heart failure and death. There are multiple molecules and signalling pathways that are involved in the vascular remodelling, including non-coding RNAs, i.e. microRNAs and long non-coding RNAs (lncRNAs). It is only in recent years that the role of lncRNAs in the pathobiology of pulmonary vascular remodelling and right ventricular dysfunction is being vigorously investigated. In this review, we have summarized the current state of knowledge about the role of lncRNAs as key drivers and gatekeepers in regulating major cellular and molecular trafficking involved in the pathogenesis of PAH. In addition, we have discussed the limitations and challenges in translating lncRNA research in vivo and in therapeutic applications of lncRNAs in PAH.
Collapse
Affiliation(s)
- Kashif Rafiq Zahid
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
- Key Laboratory of Optoelectronic Devices, Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Umar Raza
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Khadim Abid Majeed Road, Rawalpindi, Pakistan
| | - Jidong Chen
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
| | - Usha J Raj
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, USA
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
| |
Collapse
|
132
|
He M, Martin M, Marin T, Chen Z, Gongol B. Endothelial mechanobiology. APL Bioeng 2020; 4:010904. [PMID: 32095737 PMCID: PMC7032971 DOI: 10.1063/1.5129563] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/19/2020] [Indexed: 12/11/2022] Open
Abstract
Lining the luminal surface of the vasculature, endothelial cells (ECs) are in direct
contact with and differentially respond to hemodynamic forces depending on their anatomic
location. Pulsatile shear stress (PS) is defined by laminar flow and is predominantly
located in straight vascular regions, while disturbed or oscillatory shear stress (OS) is
localized to branch points and bifurcations. Such flow patterns have become a central
focus of vascular diseases, such as atherosclerosis, because the focal distribution of
endothelial dysfunction corresponds to regions exposed to OS, whereas endothelial
homeostasis is maintained in regions defined by PS. Deciphering the mechanotransduction
events that occur in ECs in response to differential flow patterns has required the
innovation of multidisciplinary approaches in both in vitro and
in vivo systems. The results from these studies have identified a
multitude of shear stress-regulated molecular networks in the endothelium that are
implicated in health and disease. This review outlines the significance of scientific
findings generated in collaboration with Dr. Shu Chien.
Collapse
Affiliation(s)
- Ming He
- Department of Medicine, University of California, San Diego, California 92093, USA
| | - Marcy Martin
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Traci Marin
- Department of Health Sciences, Victor Valley College, Victorville, California 92395, USA
| | - Zhen Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, California 91010, USA
| | - Brendan Gongol
- Department of Medicine, University of California, San Diego, California 92093, USA
| |
Collapse
|
133
|
Steinberg GR, Carling D. AMP-activated protein kinase: the current landscape for drug development. Nat Rev Drug Discov 2020; 18:527-551. [PMID: 30867601 DOI: 10.1038/s41573-019-0019-2] [Citation(s) in RCA: 401] [Impact Index Per Article: 100.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since the discovery of AMP-activated protein kinase (AMPK) as a central regulator of energy homeostasis, many exciting insights into its structure, regulation and physiological roles have been revealed. While exercise, caloric restriction, metformin and many natural products increase AMPK activity and exert a multitude of health benefits, developing direct activators of AMPK to elicit beneficial effects has been challenging. However, in recent years, direct AMPK activators have been identified and tested in preclinical models, and a small number have entered clinical trials. Despite these advances, which disease(s) represent the best indications for therapeutic AMPK activation and the long-term safety of such approaches remain to be established.
Collapse
Affiliation(s)
- Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - David Carling
- Cellular Stress Group, Medical Research Council London Institute of Medical Sciences, Hammersmith Hospital, Imperial College, London, UK
| |
Collapse
|
134
|
Liu J, Li X, Lu Q, Ren D, Sun X, Rousselle T, Li J, Leng J. AMPK: a balancer of the renin-angiotensin system. Biosci Rep 2019; 39:BSR20181994. [PMID: 31413168 PMCID: PMC6722492 DOI: 10.1042/bsr20181994] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 07/24/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
The renin-angiotensin system (RAS) is undisputedly well-studied as one of the oldest and most critical regulators for arterial blood pressure, fluid volume, as well as renal function. In recent studies, RAS has also been implicated in the development of obesity, diabetes, hyperlipidemia, and other diseases, and also involved in the regulation of several signaling pathways such as proliferation, apoptosis and autophagy, and insulin resistance. AMP-activated protein kinase (AMPK), an essential cellular energy sensor, has also been discovered to be involved in these diseases and cellular pathways. This would imply a connection between the RAS and AMPK. Therefore, this review serves to draw attention to the cross-talk between RAS and AMPK, then summering the most recent literature which highlights AMPK as a point of balance between physiological and pathological functions of the RAS.
Collapse
Affiliation(s)
- Jia Liu
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, China
- Department of Surgery, University of South Florida, Tampa, FL 33612, U.S.A
| | - Xuan Li
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Qingguo Lu
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Di Ren
- Department of Surgery, University of South Florida, Tampa, FL 33612, U.S.A
| | - Xiaodong Sun
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Thomas Rousselle
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Ji Li
- Department of Surgery, University of South Florida, Tampa, FL 33612, U.S.A
| | - Jiyan Leng
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
135
|
Richards EM, Raizada MK. ACE2 and pACE2: A Pair of Aces for Pulmonary Arterial Hypertension Treatment? Am J Respir Crit Care Med 2019; 198:422-423. [PMID: 29634285 DOI: 10.1164/rccm.201803-0569ed] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
- Elaine M Richards
- 1 Department of Physiology and Functional Genomics University of Florida College of Medicine Gainesville, Florida
| | - Mohan K Raizada
- 1 Department of Physiology and Functional Genomics University of Florida College of Medicine Gainesville, Florida
| |
Collapse
|
136
|
AMPK breathing and oxygen supply. Respir Physiol Neurobiol 2019; 265:112-120. [DOI: 10.1016/j.resp.2018.08.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/06/2018] [Accepted: 08/31/2018] [Indexed: 01/28/2023]
|
137
|
Xu X, Shi L, Ma X, Su H, Ma G, Wu X, Ying K, Zhang R. RhoA-Rho associated kinase signaling leads to renin-angiotensin system imbalance and angiotensin converting enzyme 2 has a protective role in acute pulmonary embolism. Thromb Res 2019; 176:85-94. [PMID: 30784777 DOI: 10.1016/j.thromres.2019.02.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/31/2019] [Accepted: 02/13/2019] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Acute pulmonary embolism (APE) is a cardiovascular disease with high morbidity and mortality. Although the anatomical obstruction of the pulmonary vascular bed initiates APE, recent studies have suggested that vasoconstrictors in the renin-angiotensin system (RAS) play a role in the severity of APE. MATERIALS AND METHODS We performed a 5-year retrospective clinical study to analyze the key RAS components in APE patients, including angiotensin converting enzyme (ACE), ACE2, angiotensin II (Ang II) and angiotensin 1-7(Ang(1-7)). The role of RhoA-Rho associated kinase (ROCK) signaling in regulating RAS vasoconstrictors was detected in rat pulmonary artery endothelial cells and in an APE rat model. RESULTS In clinical study, we found that the levels of RAS vasoconstrictors were correlated with the clinical classification of APE patients, ACE and Ang II were unregulated, whereas ACE2 and Ang(1-7) were downregulated in the high-risk group compared to the healthy volunteers. In animal study, we found that activated RhoA-ROCK signaling was responsible for the imbalance in RAS vasoconstrictors both in vitro and in vivo, and further evidence indicated that ROCK inhibitors (Y27632 or HA1077) and an ACE2 activator (Resorcinol naphthalein) restored the dysregulated RAS vasoconstrictors significantly and had a protective role in an APE rat model. CONCLUSIONS Our study revealed that RhoA-ROCK signaling leads to RAS imbalance in APE patients, and ACE2 activation might be a novel therapeutic target in APE treatment.
Collapse
Affiliation(s)
- Xiaoling Xu
- Department of Pulmonary and Critical Care Medicine, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Liuhong Shi
- Department of Ultrasound, the Second Affiliated Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Xiuqing Ma
- Department of Pulmonary and Critical Care Medicine, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Hua Su
- Department of Pulmonary and Critical Care Medicine, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Guofeng Ma
- Department of Pulmonary and Critical Care Medicine, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Xiaohong Wu
- Department of Pulmonary and Critical Care Medicine, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Kejing Ying
- Department of Pulmonary and Critical Care Medicine, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Ruifeng Zhang
- Department of Pulmonary and Critical Care Medicine, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China.
| |
Collapse
|
138
|
Santos RAS. Genetic Models. ANGIOTENSIN-(1-7) 2019. [PMCID: PMC7120897 DOI: 10.1007/978-3-030-22696-1_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Genetically altered rat and mouse models have been instrumental in the functional analysis of genes in a physiological context. In particular, studies on the renin-angiotensin system (RAS) have profited from this technology in the past. In this review, we summarize the existing animal models for the protective axis of the RAS consisting of angiotensin-converting enzyme 2 (ACE2), angiotensin-(1-7)(Ang-(1-7), and its receptor Mas. With the help of models with altered expression of the components of this axis in the brain and cardiovascular organs, its physiological and pathophysiological functions have been elucidated. Thus, novel opportunities for therapeutic interventions in cardiovascular diseases were revealed targeting ACE2 or Mas.
Collapse
|
139
|
Abstract
In the present chapter, we review and summarize current advances on the role of angiotensin-(1-7) [Ang-(1-7)] in the pathophysiology of main lung diseases: pulmonary hypertension (PH), acute respiratory distress syndrome (ARDS), asthma, and pulmonary fibrosis. Understanding the involvement of renin angiotensin system (RAS) in pulmonary inflammation may open new therapeutic possibilities for the treatment of respiratory diseases. Studies to date showed that Ang-(1-7) presents anti-inflammatory, antifibrotic activities and reduces pulmonary remodeling. These actions support the development of new pharmacological therapies based on the increase in Ang-(1-7) in the lungs to improve the treatment of inflammatory diseases.
Collapse
|
140
|
Alenina N, Bader M. ACE2 in Brain Physiology and Pathophysiology: Evidence from Transgenic Animal Models. Neurochem Res 2018; 44:1323-1329. [PMID: 30443713 PMCID: PMC7089194 DOI: 10.1007/s11064-018-2679-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/15/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022]
Abstract
Angiotensin-converting enzyme 2 (ACE2) is a protein consisting of two domains, the N-terminus is a carboxypeptidase homologous to ACE and the C-terminus is homologous to collectrin and responsible for the trafficking of the neutral amino acid transporter B(0)AT1 to the plasma membrane of gut epithelial cells. The carboxypeptidase domain not only metabolizes angiotensin II to angiotensin-(1–7), but also other peptide substrates, such as apelin, kinins and morphins. In addition, the collectrin domain regulates the levels of some amino acids in the blood, in particular of tryptophan. Therefore it is of no surprise that animals with genetic alterations in the expression of ACE2 develop a diverse pattern of phenotypes ranging from hypertension, metabolic and behavioural dysfunctions, to impairments in serotonin synthesis and neurogenesis. This review summarizes the phenotypes of such animals with a particular focus on the central nervous system.
Collapse
Affiliation(s)
- Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
- Berlin Institute of Health (BIH), Berlin, Germany.
- Charité - University Medicine, Berlin, Germany.
- Institute for Biology, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
141
|
Guignabert C, de Man F, Lombès M. ACE2 as therapy for pulmonary arterial hypertension: the good outweighs the bad. Eur Respir J 2018; 51:51/6/1800848. [PMID: 29929959 DOI: 10.1183/13993003.00848-2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Christophe Guignabert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.,Faculté de Médecine, Université Paris-Sud and Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Frances de Man
- Dept of Pulmonology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Marc Lombès
- Faculté de Médecine, Université Paris-Sud and Université Paris Saclay, Le Kremlin-Bicêtre, France.,INSERM UMR_S 1185, Le Kremlin-Bicêtre, France.,Service d'Endocrinologie et des Maladies de la Reproduction, Hôpitaux Universitaires Paris-Sud, CHU Bicêtre, AH-HP, Le Kremlin Bicêtre, France
| |
Collapse
|