1
|
Ljujic B, Maksimovic N, Damnjanovic T, Novakovic I, Grk M, Gulic M, Dusanovic-Pjevic M, Popovska Jovicic B, Rakovic I, Gazdic Jankovic M, Miletic Kovacevic M, Jekic B. HIF-1A Gene Polymorphisms are Associated With Clinical and Biochemical Parameters in COVID-19 Patients in Serbian Population. Clin Nurs Res 2025; 34:153-159. [PMID: 39781996 DOI: 10.1177/10547738241308972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The hypoxia-inducible factor-1 alpha (HIF-1 alpha) is a major regulator of adaptive response to hypoxia, common in patients with severe coronavirus disease 2019 (COVID-19). In addition, HIF-1 alpha regulates the expression of the most important proteins necessary for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of cells. The study included 129 hospitalized COVID-19 patients. Genotypes of HIF-1A gene polymorphisms rs11549465 and rs2057482 were determined by the RT-PCR method. We have observed lower mean platelet counts in carriers of HIF-1A rs11549465CC genotype (p = .050) and a significant association of thrombocytopenia with rs11549465CC/rs2057482CT HIF-1A genotypes combination (p = .037) in the group of patients under the age of 40. HIF-1A rs11549465CC genotype and rs11549465CC/rs2057482CT genotype combination could be predictive markers for thrombocytopenia in COVID-19 patients. Identification of such predictive markers for severe disease may contribute to a more efficient response of health systems to the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Biljana Ljujic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Serbia
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Nela Maksimovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Serbia
| | - Tatjana Damnjanovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Serbia
| | - Ivana Novakovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Serbia
| | - Milka Grk
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Serbia
| | - Milica Gulic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Serbia
| | | | - Biljana Popovska Jovicic
- Department of Infectious diseases, Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Ivana Rakovic
- Department of Infectious diseases, Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Marina Gazdic Jankovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Serbia
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Marina Miletic Kovacevic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Serbia
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Biljana Jekic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Serbia
| |
Collapse
|
2
|
Croatt AJ, Singh RD, Grande JP, Ackerman AW, Gurley SB, Barry MA, Juncos LA, Nath KA. ACE2 deficiency protects against heme protein-induced acute kidney injury. Am J Physiol Renal Physiol 2025; 328:F676-F683. [PMID: 40131861 DOI: 10.1152/ajprenal.00061.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) exerts countervailing effects on the renin-angiotensin-aldosterone system (RAAS). ACE2 also engages the spike protein of SARS-CoV-2. ACE2 protein has been shown recently to avidly bind heme. We examined the pathobiological relevance of this heme-binding property of ACE2 by using the glycerol-induced model of heme protein-mediated AKI (HP-AKI), which is characterized by increased kidney heme content. We studied the response of ACE2-wildtype (ACE2+/y) and ACE2-deficient (ACE2-/y) mice to HP-AKI and quantitated kidney and cellular content of heme under relevant conditions. ACE2-deficient mice, compared with ACE2-wildtype mice, were significantly protected against HP-AKI as reflected by filtration markers, less histological injury, and less expression of apoptosis and ferroptosis markers. ACE2-deficient mice also evinced lesser kidney heme content and a blunted induction of HO-1. HEK293 ACE2-overexpressing cells, compared with HEK293-native cells, when exposed to heme, retained higher amounts of heme. In HP-AKI, ACE2 expression and activity were reduced, and myoglobin and heme, administered independently, reduced ACE2 expression in the otherwise intact mouse kidney. Finally, with more severe HP-AKI, the protective effect of ACE2 deficiency was attenuated. We conclude that ACE2 deficiency confers protection against HP-AKI. We suggest that this reflects the recently recognized binding of heme to ACE2, such binding serving to facilitate renal entry of heme, a known nephrotoxin. These findings uncover a novel pathway of heme-dependent acute kidney injury. This is the first demonstration of the biological relevance of chemical binding of heme by ACE2. Finally, we identify heme proteins and heme as novel determinants of ACE2 expression.NEW & NOTEWORTHY ACE2 protein binds heme, which we reasoned would promote heme entry into the kidney and, accordingly, heme protein-mediated acute kidney injury. Our findings support this hypothesis. This study is the first to demonstrate the biological relevance of ACE2-heme binding, uncover a new pathway of heme-dependent kidney injury, and identify myoglobin and heme as novel determinants of ACE2 expression. Our study explains why plasma levels of myoglobin and heme predict poor outcomes in patients with COVID-19.
Collapse
Affiliation(s)
- Anthony J Croatt
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Raman Deep Singh
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Joseph P Grande
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Allan W Ackerman
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Susan B Gurley
- Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| | - Michael A Barry
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Luis A Juncos
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States
- Fresenius Medical Care (FME) Global Medical Office and Medical Affairs, Bad Homburg vor der Höhe, Germany
| | - Karl A Nath
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
3
|
Lynch-Miller M, Lockow S, Dümmer K, Henneck T, Olmer R, Jaboreck MC, Mergani AO, Wandrey M, Branitzki-Heinemann K, Brogden G, Naim HY, Martin U, Schulz C, Talbot SR, Meurer M, Baumgärtner W, von Köckritz-Blickwede M. Characterization of 3D human pulmonary epithelial model morphology and oxygen status under normoxia and hypoxia. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119980. [PMID: 40315920 DOI: 10.1016/j.bbamcr.2025.119980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 04/05/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025]
Abstract
Infection generates localized hypoxia in affected tissue, inducing cellular survival responses and modulating inflammatory processes. Consideration of oxygen status as a parameter in in vitro infection research is therefore vital to the generation of physiologically relevant data within the 3R context. In this study, we characterize the culture morphology and oxygenation of liquid-liquid interface (LLI) permanent bronchial epithelial (Calu-3), classical air-liquid interface (cALI) Calu-3, and cALI human primary bronchial epithelial cell (hBEC) models under the normoxic conditions within standard incubators, commonly employed in in vitro work. We compare the normoxic state of these models to their hypoxic state to assess changes in the airway epithelial environment in response to oxygen deprivation, and the extent to which select hypoxia responses can be observed at the molecular level. Additional juxtapositions are drawn between Calu-3 LLI and cALI models and Calu-3 conventional monolayer (CM) and inverted air-liquid interface (iALI) models, due to their relevance for basic and specialized research, respectively. Epithelial complexity was observed to vary amongst the filter-based models, and all models were found to exhibit characteristic extracellular oxygen depletion patterns under normoxia. Importantly, the extracellular oxygen contents of Calu-3 LLI, cALI, and CM models significantly decreased during normoxic incubation. Specific hypoxia responses through stabilization of HIF-1α, HIF-2α, and/or HIF-3α and alteration of ACE2 protein levels differed in response to both culture format and cell type. Therefore, while all models examined provide valuable opportunities for in vitro exploration, variation in their morphological, physiological, and molecular characteristics necessitates careful consideration during experimental design.
Collapse
Affiliation(s)
- Maura Lynch-Miller
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Sandra Lockow
- Department of Pathology, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Katrin Dümmer
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Timo Henneck
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Research Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Mark-Christian Jaboreck
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Research Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - AhmedElmontaser O Mergani
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Madita Wandrey
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Katja Branitzki-Heinemann
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Graham Brogden
- Institute of Experimental Virology, TWINCORE, Center for Experimental and Clinical Infection Research Hannover, Hannover, Germany
| | - Hassan Y Naim
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Research Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Claudia Schulz
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Steven R Talbot
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Marita Meurer
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany.
| |
Collapse
|
4
|
Huang T, Liu S, Ma Y, Ma L, Dang Z. Selenomethionine inhibits the proliferation of hypoxia-induced pulmonary artery smooth muscle cells by inhibiting ROS and HIF-1α-ACE-AngII axis. Sci Rep 2025; 15:11746. [PMID: 40189640 PMCID: PMC11973171 DOI: 10.1038/s41598-025-95793-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Recent studies have shown that patients with pulmonary arterial hypertension (PAH) are deficient in nutrients, especially vitamins and minerals. Selenium is a strong antioxidant and there is a correlation between selenium and quality of life in patients with PAH. The purpose of this study was to research whether Selenomethionine (SeMet) can reduce the oxidative damage of pulmonary artery smooth muscle cells (PASMCs) and inhibit the proliferation of PASMCs in hypoxia, and the protective mechanism of SeMet on hypoxia-induced PASMCs. PASMCs were cultured and divided into 5 groups, normoxia group, hypoxia group, and hypoxia + SeMet group (10,20 and 40 µg/ml). It was found that cell activity was elevated and hyperproliferation was observed in the hypoxia group compared to the normoxia control group. Meanwhile, the antioxidant indexes SOD and CAT activities were reduced, T-AOC was decreased, and ROS and MDA contents were elevated in the hypoxia group. The expressions of HIF-1α, ACE, Ang II, VEGF genes and proteins in PASMCs were increased under hypoxia. And SeMet reversed the above changes in antioxidant indicators and proteins, thereby inhibiting the proliferation of PASMCs and promoting apoptosis. Our study found that SeMet may inhibit hypoxia-induced oxidative stress and proliferation in PASMCs by the ROS and HIF-1α-ACE-AngII axis.
Collapse
Affiliation(s)
- Ting Huang
- Department of Public Health, Qinghai Unversity Medical College, Xining 810016, China
| | - Shou Liu
- Department of Public Health, Qinghai Unversity Medical College, Xining 810016, China
| | - Yanting Ma
- Department of Public Health, Qinghai Unversity Medical College, Xining 810016, China
| | - Lan Ma
- Research Center for High Attitude Medicine, Qinghai University Medical College, Xining 810016, China.
| | - Zhancui Dang
- Department of Public Health, Qinghai Unversity Medical College, Xining 810016, China.
| |
Collapse
|
5
|
Yang QW, Yue CL, Chen M, Ling YY, Dong Q, Zhou YX, Cao Y, Ding YX, Zhao X, Huang H, Zhang ZH, Hu L, Xu XH. Daphnetin may protect from SARS-CoV-2 infection by reducing ACE2. Sci Rep 2024; 14:30682. [PMID: 39730426 DOI: 10.1038/s41598-024-79734-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/12/2024] [Indexed: 12/29/2024] Open
Abstract
To combat the SARS-CoV-2 pandemic, innovative prevention strategies are needed, including reducing ACE2 expression on respiratory cells. This study screened approved drugs in China for their ability to downregulate ACE2. Daphnetin (DAP) was found to significantly reduce ACE2 mRNA and protein levels in PC9 cells. DAP exerts its inhibitory effects on ACE2 expression by targeting HIF-1α and JAK2, thereby impeding the transcription of the ACE2 gene. The SARS-CoV-2 pseudovirus infection assay confirmed that DAP-treated PC9 cells exhibited decreased susceptibility to viral infection. At therapeutic doses, DAP effectively lowers ACE2 expression in the respiratory systems of mice and humans. This suggests that DAP, already approved for other conditions, could be a new preventive measure against SARS-CoV-2, offering a cost-effective and accessible way to reduce SARS-CoV-2 spread.
Collapse
Affiliation(s)
- Qian-Wen Yang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Chang-Ling Yue
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Meng Chen
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yun-Yun Ling
- Department of Chemistry, Wannan Medical College, Wuhu, 241002, China
| | - Qi Dong
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Ying-Xin Zhou
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yin Cao
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yan-Xia Ding
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Xu Zhao
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Hai Huang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Zhao-Huan Zhang
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
| | - Lei Hu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China.
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China.
| | - Xiao-Hui Xu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China.
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China.
| |
Collapse
|
6
|
Li XC, Zhu XY, Wang YY, Tong SL, Chen ZL, Lu ZY, Zhang JH, Song LL, Wang XH, Zhang C, Sun YH, Zhong CY, Su LH, Wang LX, Huang XY. Canagliflozin alleviates pulmonary hypertension by activating PPARγ and inhibiting its S225 phosphorylation. Acta Pharmacol Sin 2024; 45:1861-1878. [PMID: 38719955 PMCID: PMC11335861 DOI: 10.1038/s41401-024-01286-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/07/2024] [Indexed: 08/22/2024]
Abstract
Pulmonary hypertension (PH) is a progressive fatal disease with no cure. Canagliflozin (CANA), a novel medication for diabetes, has been found to have remarkable cardiovascular benefits. However, few studies have addressed the effect and pharmacological mechanism of CANA in the treatment of PH. Therefore, our study aimed to investigate the effect and pharmacological mechanism of CANA in treating PH. First, CANA suppressed increased pulmonary artery pressure, right ventricular hypertrophy, and vascular remodeling in both mouse and rat PH models. Network pharmacology, transcriptomics, and biological results suggested that CANA could ameliorate PH by suppressing excessive oxidative stress and pulmonary artery smooth muscle cell proliferation partially through the activation of PPARγ. Further studies demonstrated that CANA inhibited phosphorylation of PPARγ at Ser225 (a novel serine phosphorylation site in PPARγ), thereby promoting the nuclear translocation of PPARγ and increasing its ability to resist oxidative stress and proliferation. Taken together, our study not only highlighted the potential pharmacological effect of CANA on PH but also revealed that CANA-induced inhibition of PPARγ Ser225 phosphorylation increases its capacity to counteract oxidative stress and inhibits proliferation. These findings may stimulate further research and encourage future clinical trials exploring the therapeutic potential of CANA in PH treatment.
Collapse
Affiliation(s)
- Xiu-Chun Li
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Xia-Yan Zhu
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Yang-Yue Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | | | - Zhi-Li Chen
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Zi-Yi Lu
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | | | - Lan-Lan Song
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Xing-Hong Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Chi Zhang
- Wenzhou Medical University, Wenzhou, 325000, China
| | - Yi-Han Sun
- Wenzhou Medical University, Wenzhou, 325000, China
| | | | - Li-Huang Su
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Liang-Xing Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Xiao-Ying Huang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China.
| |
Collapse
|
7
|
Yan YD, Ji WX, Zhao N, Zhang JJ, Du J, Lu T, Gu WQ. Association between ACE1 and missed abortion: ACE1 promotes H2O2-induced trophoblast cell injury in vitro†. Biol Reprod 2024; 111:406-413. [PMID: 38582608 DOI: 10.1093/biolre/ioae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/31/2024] [Accepted: 03/21/2024] [Indexed: 04/08/2024] Open
Abstract
The aim of this study was to evaluate the role of angiotensin-converting enzyme 1 (ACE1) in H2O2-induced trophoblast cell injury and the potential molecular mechanisms. Oxidative stress was modeled by exposing HTR-8/SVneo cells to 200 μM H2O2. Western blot and real-time quantitative PCR methods were used to detect protein and mRNA expression level of ACE1 in chorionic villus tissue and trophoblast HTR-8/SVneo cell. Inhibition of ACE1 expression was achieved by transfection with small interfering RNA. Then flow cytometry, Cell Counting Kit-8, and Transwell assay was used to assess apoptosis, viability, and migration ability of the cells. Reactive oxygen species (ROS) were detected by fluorescent probes, and malondialdehyde (MDA), superoxide dismutase (SOD), and reduced glutathione (GSH) activities were determined by corresponding detection kits. Angiotensin-converting enzyme 1 expression was upregulated in chorionic villus tissue of patients with missed abortion (MA) compared with individuals with normal early pregnancy abortion. H2O2 induced elevated ACE1 expression in HTR-8/SVneo cells, promoted apoptosis, and inhibited cell viability and migration. Knockdown of ACE1 expression inhibited H2O2-induced effects to enhance cell viability and migration and suppress apoptosis. Additionally, H2O2 stimulation caused increased levels of ROS and MDA and decreased SOD and GSH activity in the cells, whereas knockdown of ACE1 expression led to opposite changes of these oxidative stress indicators. Moreover, knockdown of ACE1 attenuated the inhibitory effect of H2O2 on the Nrf2/HO-1 pathway. Angiotensin-converting enzyme 1 was associated with MA, and it promoted H2O2-induced injury of trophoblast cells through inhibiting the Nrf2 pathway. Therefore, ACE1 may serve as a potential therapeutic target for MA.
Collapse
Affiliation(s)
- Yi-Dan Yan
- Obstetrics and Gynecology, Changshu Hospital of Traditional Chinese Medicine, Changshu, Jiangsu, China
| | - Wen-Xia Ji
- Obstetrics and Gynecology, Changshu Hospital of Traditional Chinese Medicine, Changshu, Jiangsu, China
| | - Na Zhao
- Obstetrics and Gynecology, Changshu Hospital of Traditional Chinese Medicine, Changshu, Jiangsu, China
| | - Jian-Juan Zhang
- Obstetrics and Gynecology, Changshu Hospital of Traditional Chinese Medicine, Changshu, Jiangsu, China
| | - Jie Du
- Obstetrics and Gynecology, Changshu Hospital of Traditional Chinese Medicine, Changshu, Jiangsu, China
| | - Ting Lu
- Obstetrics and Gynecology, Changshu Hospital of Traditional Chinese Medicine, Changshu, Jiangsu, China
| | - Wei-Qun Gu
- Obstetrics and Gynecology, Changshu Hospital of Traditional Chinese Medicine, Changshu, Jiangsu, China
| |
Collapse
|
8
|
Han H, Kim JE, Lee HJ. Effect of apigetrin in pseudo-SARS-CoV-2-induced inflammatory and pulmonary fibrosis in vitro model. Sci Rep 2024; 14:14545. [PMID: 38914619 PMCID: PMC11196261 DOI: 10.1038/s41598-024-65447-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024] Open
Abstract
SARS-CoV-2 has become a global public health problem. Acute respiratory distress syndrome (ARDS) is the leading cause of death due to the SARS-CoV-2 infection. Pulmonary fibrosis (PF) is a severe and frequently reported COVID-19 sequela. In this study, an in vitro model of ARDS and PF caused by SARS-CoV-2 was established in MH-S, THP-1, and MRC-5 cells using pseudo-SARS-CoV-2 (PSCV). Expression of proinflammatory cytokines (IL-6, IL-1β, and TNF-α) and HIF-1α was increased in PSCV-infected MH-S and THP-1 cells, ARDS model, consistent with other profiling data in SARS-CoV-2-infected patients have been reported. Hypoxia-inducible factor-1 alpha (HIF-1α) siRNA and cobalt chloride were tested using this in vitro model. HIF-1α knockdown reduces inflammation caused by PSCV infection in MH-S and THP-1 cells and lowers elevated levels of CTGF, COLA1, and α-SMA in MRC-5 cells exposed to CPMSCV. Furthermore, apigetrin, a glycoside bioactive dietary flavonoid derived from several plants, including Crataegus pinnatifida, which is reported to be a HIF-1α inhibitor, was tested in this in vitro model. Apigetrin significantly reduced the increased inflammatory cytokine (IL-6, IL-1β, and TNF-α) expression and secretion by PSCV in MH-S and THP-1 cells. Apigetrin inhibited the binding of the SARS-CoV-2 spike protein RBD to the ACE2 protein. An in vitro model of PF induced by SARS-CoV-2 was produced using a conditioned medium of THP-1 and MH-S cells that were PSCV-infected (CMPSCV) into MRC-5 cells. In a PF model, CMPSCV treatment of THP-1 and MH-S cells increased cell growth, migration, and collagen synthesis in MRC-5 cells. In contrast, apigetrin suppressed the increase in cell growth, migration, and collagen synthesis induced by CMPSCV in THP-1 and MH-S MRC-5 cells. Also, compared to control, fibrosis-related proteins (CTGF, COLA1, α-SMA, and HIF-1α) levels were over two-fold higher in CMPSV-treated MRC-5 cells. Apigetrin decreased protein levels in CMPSCV-treated MRC-5 cells. Thus, our data suggest that hypoxia-inducible factor-1 alpha (HIF-1α) might be a novel target for SARS-CoV-2 sequela therapies and apigetrin, representative of HIF-1alpha inhibitor, exerts anti-inflammatory and PF effects in PSCV-treated MH-S, THP-1, and CMPVSC-treated MRC-5 cells. These findings indicate that HIF-1α inhibition and apigetrin would have a potential value in controlling SARS-CoV-2-related diseases.
Collapse
Affiliation(s)
- Hengmin Han
- Department of Cancer Preventive Material Development, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Jung-Eun Kim
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Hyo-Jeong Lee
- Department of Cancer Preventive Material Development, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
| |
Collapse
|
9
|
Pan J, Villalan AK, Ni G, Wu R, Sui S, Wu X, Wang X. Assessing eco-geographic influences on COVID-19 transmission: a global analysis. Sci Rep 2024; 14:11728. [PMID: 38777817 PMCID: PMC11111805 DOI: 10.1038/s41598-024-62300-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
COVID-19 has been massively transmitted for almost 3 years, and its multiple variants have caused serious health problems and an economic crisis. Our goal was to identify the influencing factors that reduce the threshold of disease transmission and to analyze the epidemiological patterns of COVID-19. This study served as an early assessment of the epidemiological characteristics of COVID-19 using the MaxEnt species distribution algorithm using the maximum entropy model. The transmission of COVID-19 was evaluated based on human factors and environmental variables, including climate, terrain and vegetation, along with COVID-19 daily confirmed case location data. The results of the SDM model indicate that population density was the major factor influencing the spread of COVID-19. Altitude, land cover and climatic factor showed low impact. We identified a set of practical, high-resolution, multi-factor-based maximum entropy ecological niche risk prediction systems to assess the transmission risk of the COVID-19 epidemic globally. This study provided a comprehensive analysis of various factors influencing the transmission of COVID-19, incorporating both human and environmental variables. These findings emphasize the role of different types of influencing variables in disease transmission, which could have implications for global health regulations and preparedness strategies for future outbreaks.
Collapse
Affiliation(s)
- Jing Pan
- Key Laboratory for Wildlife Diseases and Bio-Security Management of Heilongjiang Province, Heilongjiang Province, Harbin, 150040, People's Republic of China
- College of Wildlife and Protected Area, Northeast Forestry University, Heilongjiang Province, Harbin, 150040, People's Republic of China
| | - Arivizhivendhan Kannan Villalan
- Key Laboratory for Wildlife Diseases and Bio-Security Management of Heilongjiang Province, Heilongjiang Province, Harbin, 150040, People's Republic of China
- College of Wildlife and Protected Area, Northeast Forestry University, Heilongjiang Province, Harbin, 150040, People's Republic of China
| | - Guanying Ni
- HaiXi Animal Disease Control Center, Qinghai Province, Delingha, 817099, People's Republic of China
| | - Renna Wu
- HaiXi Animal Disease Control Center, Qinghai Province, Delingha, 817099, People's Republic of China
| | - ShiFeng Sui
- Zhaoyuan Forest Resources Monitoring and Protection Service Center, Shandong Province, Zhaoyuan, 265400, People's Republic of China
| | - Xiaodong Wu
- China Animal Health and Epidemiology Center, Shandong Province, Qingdao, 266032, People's Republic of China.
| | - XiaoLong Wang
- Key Laboratory for Wildlife Diseases and Bio-Security Management of Heilongjiang Province, Heilongjiang Province, Harbin, 150040, People's Republic of China.
- College of Wildlife and Protected Area, Northeast Forestry University, Heilongjiang Province, Harbin, 150040, People's Republic of China.
| |
Collapse
|
10
|
Papadopoulos KI, Papadopoulou A, Aw TC. Anexelekto (AXL) no more: microRNA-155 (miR-155) controls the "Uncontrolled" in SARS-CoV-2. Hum Cell 2024; 37:582-592. [PMID: 38472734 DOI: 10.1007/s13577-024-01041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/30/2024] [Indexed: 03/14/2024]
Abstract
AXL is the gene that encodes the Anexelekto (AXL) receptor tyrosine kinase that demonstrates significant roles in various cellular processes, including cell growth, survival, and migration. Anexelekto is a Greek word meaning excessive and uncontrolled, semantically implying the crucial involvement of AXL in cancer and immune biology, and in promoting cancer metastasis. AXL overexpression appears to drive epithelial to mesenchymal transition, tumor angiogenesis, decreased antitumor immune response, and resistance to therapeutic agents. Recently, AXL has been reported to play important roles in several viral infections, including SARS-CoV-2. We have previously outlined the importance of microRNAs (miRNAs, miRs) and especially miR-155 in SARS-CoV-2 pathophysiology through regulation of the Renin-Angiotensin Aldosterone System (RAAS) and influence on several aspects of host innate immunity. MiRNAs are negative regulators of gene expression, decreasing the stability of target RNAs or limiting their translation and, enthrallingly, miR-155 is also involved in AXL homeostasis-both endogenously and pharmaceutically using repurposed drugs (e.g., metformin)-highlighting thrifty evolutionary host innate immunity mechanisms that successfully can thwart viral entry and replication. Cancer, infections, and immune system disturbances will increasingly involve miRNA diagnostics and therapeutics in the future.
Collapse
Affiliation(s)
- K I Papadopoulos
- THAI StemLife, 566/3 Soi Ramkhamhaeng 39 (Thepleela 1), Prachaouthit Rd, Wangthonglang, Bangkok, 10310, Thailand.
| | - A Papadopoulou
- Feelgood Lund, Occupational and Environmental Health Services, Ideon Science Park, Scheelevägen 17, 223 63, Lund, Sweden
| | - T C Aw
- Department of Laboratory Medicine, Changi General Hospital, 2 Simei Street 3, Singapore, 529889, Singapore
- Department of Medicine, National University of Singapore, Singapore, 119228, Singapore
| |
Collapse
|
11
|
Cunningham JW, Claggett BL, Lopes RD, McMurray JJ, Perkovic V, Carroll K, Hiemstra T, Khavandi K, Lukas MA, Ranganathan P, Shannon J, van Adelsberg J, Singh AK, Solomon SD. Daprodustat and Heart Failure in CKD. J Am Soc Nephrol 2024; 35:607-617. [PMID: 38383961 PMCID: PMC11149037 DOI: 10.1681/asn.0000000000000321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/19/2024] [Indexed: 02/23/2024] Open
Abstract
Key Points Patients with CKD face meaningful risk of heart failure hospitalization. Daprodustat compared with darbepoetin was associated with a nonsignificantly greater number of heart failure hospitalizations in non-dialysis patients. Background Patients with CKD are at higher risk of heart failure. The hypoxia-inducible factor prolyl hydroxylase inhibitor daprodustat is an orally acting alternative to conventional injectable erythropoietin-stimulating agents (ESAs) for the treatment of anemia in patients with CKD. Whether daprodustat affects the risk of heart failure hospitalization is unknown. Methods The Anemia Studies in Chronic Kidney Disease: Erythropoiesis via a Novel Prolyl Hydroxylase Inhibitor Daprodustat–Dialysis (ASCEND-D; n =2964) and Anemia Studies in Chronic Kidney Disease: Erythropoiesis via a Novel Prolyl Hydroxylase Inhibitor Daprodustat–Non-Dialysis (ASCEND-ND; n =3872) trials compared daprodustat with conventional ESA in patients with anemia of CKD who did or did not require dialysis, respectively. We identified risk factors of heart failure hospitalization and assessed the effect of daprodustat compared with conventional ESA on heart failure hospitalizations. Results History of heart failure, diabetes, and higher systolic BP were independently associated with heart failure hospitalization in both trials, irrespective of treatment assignment. The number of first heart failure hospitalizations was greater in the daprodustat arm in patients not receiving dialysis (hazard ratio [HR], 1.22 [95% confidence interval (CI), 0.95 to 1.56], P = 0.12) and in patients receiving dialysis (HR, 1.10 [95% CI, 0.84 to 1.45], P = 0.47), although these differences were not statistically significant. HRs in patients with and without history of heart failure were 1.37 (95% CI, 0.89 to 2.11) versus 1.08 (95% CI, 0.79 to 1.46) (P -interaction=0.36) in the ASCEND-ND trial and 1.52 (95% CI, 0.97 to 2.38) versus 0.93 (95% CI, 0.66 to 1.30) (P -interaction=0.09) in the ASCEND-D trial, respectively. In post hoc analyses, daprodustat increased total (first and recurrent) heart failure hospitalizations in participants not receiving dialysis (rate ratio, 1.46 [95% CI, 1.11 to 1.92], P = 0.007) but not in participants receiving dialysis (rate ratio, 1.01 [95% CI, 0.74 to 1.39], P = 0.93). Daprodustat did not significantly affect the risk of a composite outcome of first heart failure hospitalization or death. Conclusions A greater number of first heart failure hospitalization events occurred in patients treated with daprodustat compared with conventional ESA, but this difference did not reach statistical significance. Differences in the number of heart failure hospitalization events were most apparent in patients not receiving dialysis and in patients with history of heart failure. Podcast This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/JASN/2024_04_10_ASN0000000000000321.mp3
Collapse
Affiliation(s)
| | - Brian L. Claggett
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Renato D. Lopes
- Duke University School of Medicine and Duke Clinical Research Institute, Durham, North Carolina
| | - John J.V. McMurray
- BHF Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Vlado Perkovic
- University of New South Wales, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | - Ajay K. Singh
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Scott D. Solomon
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
12
|
May L, Chu CF, Zielinski CE. Single-Cell RNA Sequencing Reveals HIF1A as a Severity-Sensitive Immunological Scar in Circulating Monocytes of Convalescent Comorbidity-Free COVID-19 Patients. Cells 2024; 13:300. [PMID: 38391913 PMCID: PMC10886588 DOI: 10.3390/cells13040300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
COVID-19, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is characterized by a wide range of clinical symptoms and a poorly predictable disease course. Although in-depth transcriptomic investigations of peripheral blood samples from COVID-19 patients have been performed, the detailed molecular mechanisms underlying an asymptomatic, mild or severe disease course, particularly in patients without relevant comorbidities, remain poorly understood. While previous studies have mainly focused on the cellular and molecular dissection of ongoing COVID-19, we set out to characterize transcriptomic immune cell dysregulation at the single-cell level at different time points in patients without comorbidities after disease resolution to identify signatures of different disease severities in convalescence. With single-cell RNA sequencing, we reveal a role for hypoxia-inducible factor 1-alpha (HIF1A) as a severity-sensitive long-term immunological scar in circulating monocytes of convalescent COVID-19 patients. Additionally, we show that circulating complexes formed by monocytes with either T cells or NK cells represent a characteristic cellular marker in convalescent COVID-19 patients irrespective of their preceding symptom severity. Together, these results provide cellular and molecular correlates of recovery from COVID-19 and could help in immune monitoring and in the design of new treatment strategies.
Collapse
Affiliation(s)
- Lilly May
- Leibniz Institute for Natural Products Research and Infection Biology, Department of Infection Immunology, 07745 Jena, Germany; (L.M.); (C.-F.C.)
- Center for Translational Cancer Research (TranslaTUM) & Institute of Virology, Technical University of Munich, 81675 Munich, Germany
| | - Chang-Feng Chu
- Leibniz Institute for Natural Products Research and Infection Biology, Department of Infection Immunology, 07745 Jena, Germany; (L.M.); (C.-F.C.)
- Center for Translational Cancer Research (TranslaTUM) & Institute of Virology, Technical University of Munich, 81675 Munich, Germany
| | - Christina E. Zielinski
- Leibniz Institute for Natural Products Research and Infection Biology, Department of Infection Immunology, 07745 Jena, Germany; (L.M.); (C.-F.C.)
- Center for Translational Cancer Research (TranslaTUM) & Institute of Virology, Technical University of Munich, 81675 Munich, Germany
- Department of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| |
Collapse
|
13
|
Curran CS, Cui X, Li Y, Jeakle M, Sun J, Demirkale CY, Minkove S, Hoffmann V, Dhamapurkar R, Chumbris S, Bolyard C, Iheanacho A, Eichacker PQ, Torabi-Parizi P. Anti-PD-L1 therapy altered inflammation but not survival in a lethal murine hepatitis virus-1 pneumonia model. Front Immunol 2024; 14:1308358. [PMID: 38259435 PMCID: PMC10801642 DOI: 10.3389/fimmu.2023.1308358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Because prior immune checkpoint inhibitor (ICI) therapy in cancer patients presenting with COVID-19 may affect outcomes, we investigated the beta-coronavirus, murine hepatitis virus (MHV)-1, in a lethal pneumonia model in the absence (Study 1) or presence of prior programmed cell death ligand-1 (PD-L1) antibody (PD-L1mAb) treatment (Study 2). Methods In Study 1, animals were inoculated intratracheally with MHV-1 or vehicle and evaluated at day 2, 5, and 10 after infection. In Study 2, uninfected or MHV-1-infected animals were pretreated intraperitoneally with control or PD-L1-blocking antibodies (PD-L1mAb) and evaluated at day 2 and 5 after infection. Each study examined survival, physiologic and histologic parameters, viral titers, lung immunophenotypes, and mediator production. Results Study 1 results recapitulated the pathogenesis of COVID-19 and revealed increased cell surface expression of checkpoint molecules (PD-L1, PD-1), higher expression of the immune activation marker angiotensin converting enzyme (ACE), but reduced detection of the MHV-1 receptor CD66a on immune cells in the lung, liver, and spleen. In addition to reduced detection of PD-L1 on all immune cells assayed, PD-L1 blockade was associated with increased cell surface expression of PD-1 and ACE, decreased cell surface detection of CD66a, and improved oxygen saturation despite reduced blood glucose levels and increased signs of tissue hypoxia. In the lung, PD-L1mAb promoted S100A9 but inhibited ACE2 production concomitantly with pAKT activation and reduced FOXO1 levels. PD-L1mAb promoted interferon-γ but inhibited IL-5 and granulocyte-macrophage colony-stimulating factor (GM-CSF) production, contributing to reduced bronchoalveolar lavage levels of eosinophils and neutrophils. In the liver, PD-L1mAb increased viral clearance in association with increased macrophage and lymphocyte recruitment and liver injury. PD-L1mAb increased the production of virally induced mediators of injury, angiogenesis, and neuronal activity that may play role in COVID-19 and ICI-related neurotoxicity. PD-L1mAb did not affect survival in this murine model. Discussion In Study 1 and Study 2, ACE was upregulated and CD66a and ACE2 were downregulated by either MHV-1 or PD-L1mAb. CD66a is not only the MHV-1 receptor but also an identified immune checkpoint and a negative regulator of ACE. Crosstalk between CD66a and PD-L1 or ACE/ACE2 may provide insight into ICI therapies. These networks may also play role in the increased production of S100A9 and neurological mediators in response to MHV-1 and/or PD-L1mAb, which warrant further study. Overall, these findings support observational data suggesting that prior ICI treatment does not alter survival in patients presenting with COVID-19.
Collapse
Affiliation(s)
- Colleen S. Curran
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xizhong Cui
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Yan Li
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Mark Jeakle
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Cumhur Y. Demirkale
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Samuel Minkove
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Victoria Hoffmann
- Division of Veterinary Resources, National Institutes of Health, Bethesda, MD, United States
| | - Rhea Dhamapurkar
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Symya Chumbris
- Texcell North-America, Inc., Frederick, MD, United States
| | | | | | - Peter Q. Eichacker
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Parizad Torabi-Parizi
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
14
|
Sivagurunathan N, Calivarathan L. SARS-CoV-2 Infection to Premature Neuronal Aging and Neurodegenerative Diseases: Is there any Connection with Hypoxia? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:431-448. [PMID: 37073650 DOI: 10.2174/1871527322666230418114446] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 01/28/2023] [Accepted: 02/09/2023] [Indexed: 04/20/2023]
Abstract
The pandemic of coronavirus disease-2019 (COVID-19), caused by SARS-CoV-2, has become a global concern as it leads to a spectrum of mild to severe symptoms and increases death tolls around the world. Severe COVID-19 results in acute respiratory distress syndrome, hypoxia, and multi- organ dysfunction. However, the long-term effects of post-COVID-19 infection are still unknown. Based on the emerging evidence, there is a high possibility that COVID-19 infection accelerates premature neuronal aging and increases the risk of age-related neurodegenerative diseases in mild to severely infected patients during the post-COVID period. Several studies correlate COVID-19 infection with neuronal effects, though the mechanism through which they contribute to the aggravation of neuroinflammation and neurodegeneration is still under investigation. SARS-CoV-2 predominantly targets pulmonary tissues and interferes with gas exchange, leading to systemic hypoxia. The neurons in the brain require a constant supply of oxygen for their proper functioning, suggesting that they are more vulnerable to any alteration in oxygen saturation level that results in neuronal injury with or without neuroinflammation. We hypothesize that hypoxia is one of the major clinical manifestations of severe SARS-CoV-2 infection; it directly or indirectly contributes to premature neuronal aging, neuroinflammation, and neurodegeneration by altering the expression of various genes responsible for the survival of the cells. This review focuses on the interplay between COVID-19 infection, hypoxia, premature neuronal aging, and neurodegenerative diseases and provides a novel insight into the molecular mechanisms of neurodegeneration.
Collapse
Affiliation(s)
- Narmadhaa Sivagurunathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur - 610005, Tamil Nadu, India
| | - Latchoumycandane Calivarathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur - 610005, Tamil Nadu, India
| |
Collapse
|
15
|
Araque-Rodriguez SA, Solarte I, Rojas-Roa N, Rodriguez-Villamizar LA. Altitude and COVID-19 in Colombia: An updated analysis accounting for potential confounders. Respir Physiol Neurobiol 2023; 316:104136. [PMID: 37532001 DOI: 10.1016/j.resp.2023.104136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/10/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
We assessed the relationship between the altitude of municipalities and the incidence, mortality, and fatality from COVID-19 and excess of mortality in Colombia between 2020 and 2022. We conducted an ecologic study including all 1122 municipalities in Colombia and used categories of altitude as main independent variable. We fit multivariable regression models for incidence, mortality, fatality rates, and excess of mortality controlling for several variables at municipality level. There was a higher incidence rate, similar mortality rate and lower case-fatality rate for COVID-19 during 2020-2022 in municipalities in the upper category of altitude (>=2500 masl) compared to the lower category (<1000 masl). The excess of mortality was lower but not statistically different in municipalities in the upper category of altitude, and significantly lower in the intermediate altitude category compared to the lowlands. Our findings provide evidence that municipalities with high altitude had similar mortality rate, and lower case-fatality rate and excess of mortality for COVID-19 compared to lowlands in Colombia.
Collapse
Affiliation(s)
- Santiago A Araque-Rodriguez
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga, Calle 157 14-55, 681001 Floridablanca, Colombia
| | - Iván Solarte
- Facultad de Medicina, Pontificia Universidad Javeriana, Carrera 7 40-62, Bogotá, Colombia; Unidad de Neumología, Hospital Universitario San Ignacio, Carrera 7 40-62, Bogotá, Colombia
| | - Néstor Rojas-Roa
- Facultad de Ingenierías, Universidad Nacional de Colombia, Edificio 401, Carrera 45 26-85, Bogotá, Colombia
| | - Laura A Rodriguez-Villamizar
- Departamento de Salud Pública, Escuela de Medicina, Universidad Industrial de Santander, Carrera 32 29-31 of 301, 68002, Bucaramanga, Colombia.
| |
Collapse
|
16
|
Jones EAV. Mechanism of COVID-19-Induced Cardiac Damage from Patient, In Vitro and Animal Studies. Curr Heart Fail Rep 2023; 20:451-460. [PMID: 37526812 PMCID: PMC10589152 DOI: 10.1007/s11897-023-00618-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 08/02/2023]
Abstract
PURPOSE OF REVIEW Though patient studies have been important for understanding the disease, research done in animals and cell culture complement our knowledge from patient data and provide insight into the mechanism of the disease. Understanding how COVID causes damage to the heart is essential to understanding possible long-term consequences. RECENT FINDINGS COVID-19 is primarily a disease that attacks the lungs; however, it is known to have important consequences in many other tissues including the heart. Though myocarditis does occur in some patients, for most cases of cardiac damage, the injury arises from scarring either due to myocardial infarction or micro-infarction. The main focus is on how COVID affects blood flow through the coronaries. We review how endothelial activation leads to a hypercoagulative state in COVID-19. We also emphasize the effects that the cytokine storm can directly have on the regulation of coronary blood flow. Since the main two cell types that can be infected in the heart are pericytes and cardiomyocytes, we further describe the known effects on pericyte function and how that can further lead to microinfarcts within the heart. Though many of these effects are systemic, this review focuses on the consequences on cardiac tissue of this dysregulation and the role that it has in the formation of myocardial scarring.
Collapse
Affiliation(s)
- Elizabeth A V Jones
- Centre for Molecular and Vascular Biology, Herestraat 49, Bus 911, 3000, KU, Leuven, Belgium.
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, Netherlands.
| |
Collapse
|
17
|
Wang CW, Chuang HC, Tan TH. ACE2 in chronic disease and COVID-19: gene regulation and post-translational modification. J Biomed Sci 2023; 30:71. [PMID: 37608279 PMCID: PMC10464117 DOI: 10.1186/s12929-023-00965-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/15/2023] [Indexed: 08/24/2023] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2), a counter regulator of the renin-angiotensin system, provides protection against several chronic diseases. Besides chronic diseases, ACE2 is the host receptor for SARS-CoV or SARS-CoV-2 virus, mediating the first step of virus infection. ACE2 levels are regulated by transcriptional, post-transcriptional, and post-translational regulation or modification. ACE2 transcription is enhanced by transcription factors including Ikaros, HNFs, GATA6, STAT3 or SIRT1, whereas ACE2 transcription is reduced by the transcription factor Brg1-FoxM1 complex or ERRα. ACE2 levels are also regulated by histone modification or miRNA-induced destabilization. The protein kinase AMPK, CK1α, or MAP4K3 phosphorylates ACE2 protein and induces ACE2 protein levels by decreasing its ubiquitination. The ubiquitination of ACE2 is induced by the E3 ubiquitin ligase MDM2 or UBR4 and decreased by the deubiquitinase UCHL1 or USP50. ACE2 protein levels are also increased by the E3 ligase PIAS4-mediated SUMOylation or the methyltransferase PRMT5-mediated ACE2 methylation, whereas ACE2 protein levels are decreased by AP2-mediated lysosomal degradation. ACE2 is downregulated in several human chronic diseases like diabetes, hypertension, or lung injury. In contrast, SARS-CoV-2 upregulates ACE2 levels, enhancing host cell susceptibility to virus infection. Moreover, soluble ACE2 protein and exosomal ACE2 protein facilitate SARS-CoV-2 infection into host cells. In this review, we summarize the gene regulation and post-translational modification of ACE2 in chronic disease and COVID-19. Understanding the regulation and modification of ACE2 may help to develop prevention or treatment strategies for ACE2-mediated diseases.
Collapse
Affiliation(s)
- Chia-Wen Wang
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053 Taiwan
| | - Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053 Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053 Taiwan
| |
Collapse
|
18
|
Akinsulie OC, Shahzad S, Ogunleye SC, Oladapo IP, Joshi M, Ugwu CE, Gbadegoye JO, Hassan FO, Adeleke R, Afolabi Akande Q, Adesola RO. Crosstalk between hypoxic cellular micro-environment and the immune system: a potential therapeutic target for infectious diseases. Front Immunol 2023; 14:1224102. [PMID: 37600803 PMCID: PMC10434535 DOI: 10.3389/fimmu.2023.1224102] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/26/2023] [Indexed: 08/22/2023] Open
Abstract
There are overwhelming reports on the promotional effect of hypoxia on the malignant behavior of various forms of cancer cells. This has been proposed and tested exhaustively in the light of cancer immunotherapy. However, there could be more interesting functions of a hypoxic cellular micro-environment than malignancy. There is a highly intricate crosstalk between hypoxia inducible factor (HIF), a transcriptional factor produced during hypoxia, and nuclear factor kappa B (NF-κB) which has been well characterized in various immune cell types. This important crosstalk shares common activating and inhibitory stimuli, regulators, and molecular targets. Impaired hydroxylase activity contributes to the activation of HIFs. Inflammatory ligands activate NF-κB activity, which leads to the expression of inflammatory and anti-apoptotic genes. The eventual sequelae of the interaction between these two molecular players in immune cells, either bolstering or abrogating functions, is largely cell-type dependent. Importantly, this holds promise for interesting therapeutic interventions against several infectious diseases, as some HIF agonists have helped prevent immune-related diseases. Hypoxia and inflammation are common features of infectious diseases. Here, we highlighted the role of this crosstalk in the light of functional immunity against infection and inflammation, with special focus on various innate and adaptive immune cells. Particularly, we discussed the bidirectional effects of this crosstalk in the regulation of immune responses by monocytes/macrophages, dendritic cells, neutrophils, B cells, and T cells. We believe an advanced understanding of the interplay between HIFs and NF-kB could reveal novel therapeutic targets for various infectious diseases with limited treatment options.
Collapse
Affiliation(s)
- Olalekan Chris Akinsulie
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Sammuel Shahzad
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Seto Charles Ogunleye
- College of Veterinary Medicine, Mississippi State University, Starkville, MS, United States
| | - Ifeoluwa Peace Oladapo
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Melina Joshi
- Center for Molecular Dynamics Nepal, Kathmandu, Nepal
| | - Charles Egede Ugwu
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, United States
| | - Joy Olaoluwa Gbadegoye
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Fasilat Oluwakemi Hassan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Richard Adeleke
- College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Qudus Afolabi Akande
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | | |
Collapse
|
19
|
Mia MS, Hossain D, Woodbury E, Kelleher S, Palamuttam RJ, Rao R, Steen P, Jarajapu YP, Mathew S. Integrin β1 is a key determinant of the expression of angiotensin-converting enzyme 2 (ACE2) in the kidney epithelial cells. Eur J Cell Biol 2023; 102:151316. [PMID: 37084657 PMCID: PMC11086052 DOI: 10.1016/j.ejcb.2023.151316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023] Open
Abstract
The expression of the angiotensin-converting enzyme 2 (ACE2) is altered in multiple chronic kidney diseases like hypertension and renal fibrosis, where the signaling from the basal membrane proteins is critical for the development and progression of the various pathologies. Integrins are heterodimeric cell surface receptors that have important roles in the progression of these chronic kidney diseases by altering various cell signaling pathways in response to changes in the basement membrane proteins. It is unclear whether integrin or integrin-mediated signaling affects the ACE2 expression in the kidney. The current study tests the hypothesis that integrin β1 regulates the expression of ACE2 in kidney epithelial cells. The role of integrin β1 in ACE2 expression in renal epithelial cells was investigated by shRNA-mediated knockdown and pharmacological inhibition. In vivo studies were carried out using epithelial cell-specific deletion of integrin β1 in the kidneys. Deletion of integrin β1 from the mouse renal epithelial cells reduced the expression of ACE2 in the kidney. Furthermore, the downregulation of integrin β1 using shRNA decreased ACE2 expression in human renal epithelial cells. ACE2 expression levels were also decreased in renal epithelial cells and cancer cells when treated with an integrin α2β1 antagonist, BTT 3033. SARS-CoV-2 viral entry to human renal epithelial cells and cancer cells was also inhibited by BTT 3033. This study demonstrates that integrin β1 positively regulates the expression of ACE2, which is required for the entry of SARS-CoV-2 into kidney cells.
Collapse
Affiliation(s)
- Md Saimon Mia
- Department of Pharmaceutical Sciences, School of Pharmacy, North Dakota State University, Fargo, ND, USA
| | - Delowar Hossain
- Department of Pharmaceutical Sciences, School of Pharmacy, North Dakota State University, Fargo, ND, USA
| | - Emerson Woodbury
- Department of Pharmaceutical Sciences, School of Pharmacy, North Dakota State University, Fargo, ND, USA
| | - Sean Kelleher
- Department of Pharmaceutical Sciences, School of Pharmacy, North Dakota State University, Fargo, ND, USA
| | | | - Reena Rao
- Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Preston Steen
- Sanford Health Roger Maris Cancer Center, Fargo, ND, USA
| | - Yagna Pr Jarajapu
- Department of Pharmaceutical Sciences, School of Pharmacy, North Dakota State University, Fargo, ND, USA
| | - Sijo Mathew
- Department of Pharmaceutical Sciences, School of Pharmacy, North Dakota State University, Fargo, ND, USA; Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
20
|
Kuo CW, Su PL, Huang TH, Lin CC, Chen CW, Tsai JS, Liao XM, Chan TY, Shieh CC. Cigarette smoke increases susceptibility of alveolar macrophages to SARS-CoV-2 infection through inducing reactive oxygen species-upregulated angiotensin-converting enzyme 2 expression. Sci Rep 2023; 13:7894. [PMID: 37193781 DOI: 10.1038/s41598-023-34785-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/08/2023] [Indexed: 05/18/2023] Open
Abstract
Alveolar macrophages (AMs) are the drivers of pulmonary cytokine storm in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This study aimed to investigate clinical-regulatory factors for the entrance protein of SARS-CoV-2, angiotensin-converting enzyme 2 (ACE2) in AMs. Human AMs were collected from 56 patients using bronchoalveolar lavage. ACE2 expression in AMs was positively correlated with smoking pack-year (Spearman's r = 0.347, P = 0.038). In multivariate analysis, current smoking was associated with increased ACE2 in AMs (β-coefficient: 0.791, 95% CI 0.019-1.562, P = 0.045). In vitro study, ex-vivo human AMs with higher ACE2 were more susceptible to SARS-CoV-2 pseudovirus (CoV-2 PsV). Treating human AMs using cigarette smoking extract (CSE) increases the ACE2 and susceptibility to CoV-2 PsV. CSE did not significantly increase the ACE2 in AMs of reactive oxygen species (ROS) deficient Cybb-/- mice; however, exogenous ROS increased the ACE2 in Cybb-/- AMs. N-acetylcysteine (NAC) decreases ACE2 by suppressing intracellular ROS in human AMs. In conclusion, cigarette smoking increases the susceptibility to SARS-CoV-2 by increasing ROS-induced ACE2 expression of AMs. Further investigation into the preventive effect of NAC on the pulmonary complications of COVID-19 is required.
Collapse
Affiliation(s)
- Chin-Wei Kuo
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70403, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Lan Su
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tang-Hsiu Huang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70403, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Chung Lin
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chian-Wei Chen
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jeng-Shiuan Tsai
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Xin-Min Liao
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70403, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Yi Chan
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Chang Shieh
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70403, Taiwan.
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
21
|
Fricke-Galindo I, Buendia-Roldan I, Ponce-Aguilar DI, Pérez-Rubio G, Chavez-Galan L, Alanis-Ponce J, Pérez-Torres K, Valencia-Pérez Rea D, Téllez-Quijada F, Nava-Quiroz KJ, Hernández-Zenteno RDJ, Gutiérrez-Nava A, Falfán-Valencia R. The ACE rs1799752 Variant Is Associated with COVID-19 Severity but Is Independent of Serum ACE Activity in Hospitalized and Recovered Patients. Int J Mol Sci 2023; 24:7678. [PMID: 37108839 PMCID: PMC10142321 DOI: 10.3390/ijms24087678] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
This paper assesses the association of the insertion/deletion ACE (angiotensin-converting enzyme) variant (rs1799752 I/D) and the serum ACE activity with the severity of COVID-19 as well as its impact on post-COVID-19, and we compare these associations with those for patients with non-COVID-19 respiratory disorders. We studied 1252 patients with COVID-19, 104 subjects recovered from COVID-19, and 74 patients hospitalized with a respiratory disease different from COVID-19. The rs1799752 ACE variant was assessed using TaqMan® Assays. The serum ACE activity was determined using a colorimetric assay. The DD genotype was related to risk for invasive mechanical ventilation (IMV) requirement as an indicator of COVID-19 severity when compared to the frequencies of II + ID genotypes (p = 0.025, OR = 1.428, 95% CI = 1.046-1.949). In addition, this genotype was significantly higher in COVID-19 and post-COVID-19 groups than in the non-COVID-19 subjects. The serum ACE activity levels were lower in the COVID-19 group (22.30 U/L (13.84-32.23 U/L)), which was followed by the non-COVID-19 (27.94 U/L (20.32-53.36 U/L)) and post-COVID-19 subjects (50.00 U/L (42.16-62.25 U/L)). The DD genotype of the rs1799752 ACE variant was associated with the IMV requirement in patients with COVID-19, and low serum ACE activity levels could be related to patients with severe disease.
Collapse
Affiliation(s)
- Ingrid Fricke-Galindo
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.F.-G.); (D.I.P.-A.); (G.P.-R.); (J.A.-P.); (D.V.-P.R.); (K.J.N.-Q.)
| | - Ivette Buendia-Roldan
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.B.-R.); (K.P.-T.); (F.T.-Q.)
| | - Daniel I. Ponce-Aguilar
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.F.-G.); (D.I.P.-A.); (G.P.-R.); (J.A.-P.); (D.V.-P.R.); (K.J.N.-Q.)
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico;
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.F.-G.); (D.I.P.-A.); (G.P.-R.); (J.A.-P.); (D.V.-P.R.); (K.J.N.-Q.)
| | - Leslie Chavez-Galan
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Jesús Alanis-Ponce
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.F.-G.); (D.I.P.-A.); (G.P.-R.); (J.A.-P.); (D.V.-P.R.); (K.J.N.-Q.)
| | - Karina Pérez-Torres
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.B.-R.); (K.P.-T.); (F.T.-Q.)
| | - Daniela Valencia-Pérez Rea
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.F.-G.); (D.I.P.-A.); (G.P.-R.); (J.A.-P.); (D.V.-P.R.); (K.J.N.-Q.)
| | - Fernanda Téllez-Quijada
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.B.-R.); (K.P.-T.); (F.T.-Q.)
| | - Karol J. Nava-Quiroz
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.F.-G.); (D.I.P.-A.); (G.P.-R.); (J.A.-P.); (D.V.-P.R.); (K.J.N.-Q.)
| | | | - Angélica Gutiérrez-Nava
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico;
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.F.-G.); (D.I.P.-A.); (G.P.-R.); (J.A.-P.); (D.V.-P.R.); (K.J.N.-Q.)
| |
Collapse
|
22
|
Devaux CA, Lagier JC. Unraveling the Underlying Molecular Mechanism of 'Silent Hypoxia' in COVID-19 Patients Suggests a Central Role for Angiotensin II Modulation of the AT1R-Hypoxia-Inducible Factor Signaling Pathway. J Clin Med 2023; 12:jcm12062445. [PMID: 36983445 PMCID: PMC10056466 DOI: 10.3390/jcm12062445] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
A few days after being infected with SARS-CoV-2, a fraction of people remain asymptomatic but suffer from a decrease in arterial oxygen saturation in the absence of apparent dyspnea. In light of our clinical investigation on the modulation of molecules belonging to the renin angiotensin system (RAS) in COVID-19 patients, we propose a model that explains 'silent hypoxia'. The RAS imbalance caused by SARS-CoV-2 results in an accumulation of angiotensin 2 (Ang II), which activates the angiotensin 2 type 1 receptor (AT1R) and triggers a harmful cascade of intracellular signals leading to the nuclear translocation of the hypoxia-inducible factor (HIF)-1α. HIF-1α transactivates many genes including the angiotensin-converting enzyme 1 (ACE1), while at the same time, ACE2 is downregulated. A growing number of cells is maintained in a hypoxic condition that is self-sustained by the presence of the virus and the ACE1/ACE2 ratio imbalance. This is associated with a progressive worsening of the patient's biological parameters including decreased oxygen saturation, without further clinical manifestations. When too many cells activate the Ang II-AT1R-HIF-1α axis, there is a 'hypoxic spillover', which marks the tipping point between 'silent' and symptomatic hypoxia in the patient. Immediate ventilation is required to prevent the 'hypoxic spillover'.
Collapse
Affiliation(s)
- Christian Albert Devaux
- Institut de Recherche pour le Développement, Assistance Publique Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infection Laboratory, Aix-Marseille University, 13000 Marseille, France
- Institut Hospitalo-Universitaire-Méditerranée Infection, 13000 Marseille, France
- Centre National de la Recherche Scientifique, 13000 Marseille, France
| | - Jean-Christophe Lagier
- Institut de Recherche pour le Développement, Assistance Publique Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infection Laboratory, Aix-Marseille University, 13000 Marseille, France
- Institut Hospitalo-Universitaire-Méditerranée Infection, 13000 Marseille, France
| |
Collapse
|
23
|
Motta CS, Torices S, da Rosa BG, Marcos AC, Alvarez-Rosa L, Siqueira M, Moreno-Rodriguez T, Matos ADR, Caetano BC, Martins JSCDC, Gladulich L, Loiola E, Bagshaw ORM, Stuart JA, Siqueira MM, Stipursky J, Toborek M, Adesse D. Human Brain Microvascular Endothelial Cells Exposure to SARS-CoV-2 Leads to Inflammatory Activation through NF-κB Non-Canonical Pathway and Mitochondrial Remodeling. Viruses 2023; 15:745. [PMID: 36992454 PMCID: PMC10056985 DOI: 10.3390/v15030745] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/10/2023] [Accepted: 03/03/2023] [Indexed: 03/15/2023] Open
Abstract
Neurological effects of COVID-19 and long-COVID-19, as well as neuroinvasion by SARS-CoV-2, still pose several questions and are of both clinical and scientific relevance. We described the cellular and molecular effects of the human brain microvascular endothelial cells (HBMECs) in vitro exposure by SARS-CoV-2 to understand the underlying mechanisms of viral transmigration through the blood-brain barrier. Despite the low to non-productive viral replication, SARS-CoV-2-exposed cultures displayed increased immunoreactivity for cleaved caspase-3, an indicator of apoptotic cell death, tight junction protein expression, and immunolocalization. Transcriptomic profiling of SARS-CoV-2-challenged cultures revealed endothelial activation via NF-κB non-canonical pathway, including RELB overexpression and mitochondrial dysfunction. Additionally, SARS-CoV-2 led to altered secretion of key angiogenic factors and to significant changes in mitochondrial dynamics, with increased mitofusin-2 expression and increased mitochondrial networks. Endothelial activation and remodeling can further contribute to neuroinflammatory processes and lead to further BBB permeability in COVID-19.
Collapse
Affiliation(s)
- Carolline Soares Motta
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Barbara Gomes da Rosa
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Anne Caroline Marcos
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Liandra Alvarez-Rosa
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, UFRJ, Rio de Janeiro 05508-000, Brazil
| | - Michele Siqueira
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, UFRJ, Rio de Janeiro 05508-000, Brazil
| | - Thaidy Moreno-Rodriguez
- Urology Department, University of California San Francisco, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Aline da Rocha Matos
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais (LVRE), Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Braulia Costa Caetano
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais (LVRE), Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Jessica Santa Cruz de Carvalho Martins
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais (LVRE), Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Luis Gladulich
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Erick Loiola
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Olivia R. M. Bagshaw
- Faculty of Mathematics & Science, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Jeffrey A. Stuart
- Faculty of Mathematics & Science, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Marilda M. Siqueira
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais (LVRE), Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Joice Stipursky
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, UFRJ, Rio de Janeiro 05508-000, Brazil
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daniel Adesse
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
24
|
Fevereiro-Martins M, Marques-Neves C, Guimarães H, Bicho M. Retinopathy of prematurity: A review of pathophysiology and signaling pathways. Surv Ophthalmol 2023; 68:175-210. [PMID: 36427559 DOI: 10.1016/j.survophthal.2022.11.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022]
Abstract
Retinopathy of prematurity (ROP) is a vasoproliferative disorder of the retina and a leading cause of visual impairment and childhood blindness worldwide. The disease is characterized by an early stage of retinal microvascular degeneration, followed by neovascularization that can lead to subsequent retinal detachment and permanent visual loss. Several factors play a key role during the different pathological stages of the disease. Oxidative and nitrosative stress and inflammatory processes are important contributors to the early stage of ROP. Nitric oxide synthase and arginase play important roles in ischemia/reperfusion-induced neurovascular degeneration. Destructive neovascularization is driven by mediators of the hypoxia-inducible factor pathway, such as vascular endothelial growth factor and metabolic factors (succinate). The extracellular matrix is involved in hypoxia-induced retinal neovascularization. Vasorepulsive molecules (semaphorin 3A) intervene preventing the revascularization of the avascular zone. This review focuses on current concepts about signaling pathways and their mediators, involved in the pathogenesis of ROP, highlighting new potentially preventive and therapeutic modalities. A better understanding of the intricate molecular mechanisms underlying the pathogenesis of ROP should allow the development of more effective and targeted therapeutic agents to reduce aberrant vasoproliferation and facilitate physiological retinal vascular development.
Collapse
Affiliation(s)
- Mariza Fevereiro-Martins
- Laboratório de Genética and Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Investigação Científica Bento da Rocha Cabral, Lisboa, Portugal; Departamento de Oftalmologia, Hospital Cuf Descobertas, Lisboa, Portugal.
| | - Carlos Marques-Neves
- Centro de Estudos das Ci.¼ncias da Visão, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| | - Hercília Guimarães
- Departamento de Ginecologia-Obstetrícia e Pediatria, Faculdade de Medicina, Universidade do Porto, Porto, Portugal.
| | - Manuel Bicho
- Laboratório de Genética and Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Investigação Científica Bento da Rocha Cabral, Lisboa, Portugal.
| |
Collapse
|
25
|
Aslan ES, Aydın H, Tekin YK, Keleş S, White KN, Hekim N. Association between iron metabolism and SARS-COV-2 infection, determined by ferritin, hephaestin and hypoxia-induced factor-1 alpha levels in COVID-19 patients. Mol Biol Rep 2023; 50:2471-2478. [PMID: 36600108 PMCID: PMC9812738 DOI: 10.1007/s11033-022-08221-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Due to the growing evidence of the importance of iron status in immune responses, the biomarkers of iron metabolism are of interest in novel Coronavirus Disease 2019 (COVID-19). The present prospective study was carried out to compare iron status indicated by levels of ferritin with the levels of two novel biomarkers related to iron homeostasis, hephaestin and hypoxia-inducible factors-1 (HIF-1α) in the serum of patients with COVID-19 in comparison with a control group. METHODS AND RESULTS Blood samples from 34 COVID-19 patients and from 43 healthy volunteers were collected and the levels of HEPH and HIF-1α were measured by ELISA and compared with levels of serum ferritin. COVID-19 patients had higher serum levels of ferritin than those levels in control group (P < 0.0001). Conversely levels of HIF-1α and HEPH in the COVID-19 group were significantly lower than those of control group (P < 0.0001 for both). An inverse correlation between hephaestin and ferritin as well as between HIF-1α and ferritin was found among all subjects (P < 0.0001), and among COVID-19 patients, but not to statistical significance. CONCLUSION Levels of hephaestin and HIF-1α were found to be inversely related levels of ferritin across all participants in the study, and to our knowledge this is the first report of hephaestin and HIF-1α as potential markers of iron status. Further studies are needed to corroborate the findings, utilizing a broader range of markers to monitor inflammatory as well as iron status.
Collapse
Affiliation(s)
- Elif Sibel Aslan
- Molecular Biology and Genetic Department, Faculty of Engineering and Natural Sciences, Biruni University, 10. Yıl Street. Protokol Road, Topkapı, Istanbul, Turkey.
| | - Hüseyin Aydın
- Medical Biochemistry, Department of Basic Medical Sciences, Sivas Cumhuriyet University, Sivas, Turkey
| | - Yusuf Kenan Tekin
- Medical Biochemistry, Department of Basic Medical Sciences, Sivas Cumhuriyet University, Sivas, Turkey
| | - Sami Keleş
- Ahenk Medical Diagnosis and Research Laboratory, Yeditepe University, Istanbul, Turkey
| | - Kenneth N White
- Molecular Systems for Health Research Group, School of Human Sciences, London Metropolitan University, London, UK
| | - Nezih Hekim
- Molecular Biology and Genetic Department, Faculty of Engineering and Natural Sciences, Biruni University, 10. Yıl Street. Protokol Road, Topkapı, Istanbul, Turkey
| |
Collapse
|
26
|
Exploring the Role of ACE2 as a Connecting Link between COVID-19 and Parkinson's Disease. Life (Basel) 2023; 13:life13020536. [PMID: 36836893 PMCID: PMC9961012 DOI: 10.3390/life13020536] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/30/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is frequently accompanied by neurological manifestations such as headache, delirium, and epileptic seizures, whereas ageusia and anosmia may appear before respiratory symptoms. Among the various neurological COVID-19-related comorbidities, Parkinson's disease (PD) has gained increasing attention. Some cases of PD disease have been linked to COVID-19, and both motor and non-motor symptoms in Parkinson's disease patients frequently worsen following SARS-CoV-2 infection. Although it is still unclear whether PD increases the susceptibility to SARS-CoV-2 infection or whether COVID-19 increases the risk of or unmasks future cases of PD, emerging evidence sheds more light on the molecular mechanisms underlying the relationship between these two diseases. Among them, angiotensin-converting enzyme 2 (ACE2), a significant component of the renin-angiotensin system (RAS), seems to play a pivotal role. ACE2 is required for the entry of SARS-CoV-2 to the human host cells, and ACE2 dysregulation is implicated in the severity of COVID-19-related acute respiratory distress syndrome (ARDS). ACE2 imbalance is implicated in core shared pathophysiological mechanisms between PD and COVID-19, including aberrant inflammatory responses, oxidative stress, mitochondrial dysfunction, and immune dysregulation. ACE2 may also be implicated in alpha-synuclein-induced dopaminergic degeneration, gut-brain axis dysregulation, blood-brain axis disruption, autonomic dysfunction, depression, anxiety, and hyposmia, which are key features of PD.
Collapse
|
27
|
Hypoxic pulmonary vascular response can screen subclinical lifestyle disease in healthy population. Microvasc Res 2023; 145:104454. [PMID: 36347299 DOI: 10.1016/j.mvr.2022.104454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/18/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Subclinical life style disease can cause endothelial dysfunction associated with perfusion abnormalities and reduced vascular compliance. Subclinical elevated beta type natriuretic peptide (BNP) has been associated with altered fluid shift from extra to intracellular space during acute hypoxia. Therefore we measured vascular response and BNP levels during acute hypoxia to study endothelial functions among healthy individuals. METHODS Individuals were exposed to acute normobaric hypoxia of FiO2 = 0.15 for one hour in supine position and their pulmonary and systemic vascular response to hypoxia was compared. Individuals were divided into two groups based on either no response (Group 1) or rise in systolic pulmonary artery pressure to hypoxia (Group 2) and their BNP levels were compared. RESULTS BNP was raised after hypoxia exposure in group 2 only from 18.52 ± 7 to 21.56 ± 10.82 picogram/ml, p < 0.05. Group 2 also showed an increase in mean arterial pressure and no fall in total body water in response to acute hypoxia indicating decreased endothelial function compared to Group 1. CONCLUSION Rise in pulmonary artery pressure and BNP level in response to acute normobaric hypoxia indicates reduced endothelial function and can be used to screen subclinical lifestyle disease among healthy population.
Collapse
|
28
|
Kashyap MK, Bhat A, Janjua D, Rao R, Thakur K, Chhokar A, Aggarwal N, Yadav J, Tripathi T, Chaudhary A, Senrung A, Chandra Bharti A. Role of angiotensin in different malignancies. ANGIOTENSIN 2023:505-544. [DOI: 10.1016/b978-0-323-99618-1.00019-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
29
|
Kato Y, Nishiyama K, Man Lee J, Ibuki Y, Imai Y, Noda T, Kamiya N, Kusakabe T, Kanda Y, Nishida M. TRPC3-Nox2 Protein Complex Formation Increases the Risk of SARS-CoV-2 Spike Protein-Induced Cardiomyocyte Dysfunction through ACE2 Upregulation. Int J Mol Sci 2022; 24:ijms24010102. [PMID: 36613540 PMCID: PMC9820218 DOI: 10.3390/ijms24010102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Myocardial damage caused by the newly emerged coronavirus (SARS-CoV-2) infection is one of the key determinants of COVID-19 severity and mortality. SARS-CoV-2 entry to host cells is initiated by binding with its receptor, angiotensin-converting enzyme (ACE) 2, and the ACE2 abundance is thought to reflect the susceptibility to infection. Here, we report that ibudilast, which we previously identified as a potent inhibitor of protein complex between transient receptor potential canonical (TRPC) 3 and NADPH oxidase (Nox) 2, attenuates the SARS-CoV-2 spike glycoprotein pseudovirus-evoked contractile and metabolic dysfunctions of neonatal rat cardiomyocytes (NRCMs). Epidemiologically reported risk factors of severe COVID-19, including cigarette sidestream smoke (CSS) and anti-cancer drug treatment, commonly upregulate ACE2 expression level, and these were suppressed by inhibiting TRPC3-Nox2 complex formation. Exposure of NRCMs to SARS-CoV-2 pseudovirus, as well as CSS and doxorubicin (Dox), induces ATP release through pannexin-1 hemi-channels, and this ATP release potentiates pseudovirus entry to NRCMs and human iPS cell-derived cardiomyocytes (hiPS-CMs). As the pseudovirus entry followed by production of reactive oxygen species was attenuated by inhibiting TRPC3-Nox2 complex in hiPS-CMs, we suggest that TRPC3-Nox2 complex formation triggered by panexin1-mediated ATP release participates in exacerbation of myocardial damage by amplifying ACE2-dependent SARS-CoV-2 entry.
Collapse
Affiliation(s)
- Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kazuhiro Nishiyama
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Jae Man Lee
- Laboratory of Creative Science for Insect Industries, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | - Yuko Ibuki
- Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Yumiko Imai
- Laboratory of Regulation for Intractable Infectious Diseases, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation Health and Nutrition (NIBIOHN), Osaka 567-0085, Japan
| | - Takamasa Noda
- Department of Psychiatry, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan
- Department of Neuropsychopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo 187-8553, Japan
- Department of Brain Bioregulatory Science, The Jikei University Graduate School of Medicine, Tokyo 105-8461, Japan
| | - Noriho Kamiya
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, Fukuoka 819-0395, Japan
- Division of Biotechnology, Center for Future Chemistry, Kyushu University, Fukuoka 819-0395, Japan
| | - Takahiro Kusakabe
- Laboratory of Insect Genome Science, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences (NIHS), Kawasaki 210-9501, Japan
| | - Motohiro Nishida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- National Institute for Physiological Sciences, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Correspondence: ; Tel./Fax: +81-92-642-6556
| |
Collapse
|
30
|
Significance of Catecholamine Biosynthetic/Metabolic Pathway in SARS-CoV-2 Infection and COVID-19 Severity. Cells 2022; 12:cells12010012. [PMID: 36611805 PMCID: PMC9818320 DOI: 10.3390/cells12010012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
The SARS-CoV-2 infection was previously associated with the expression of the dopamine biosynthetic enzyme L-Dopa decarboxylase (DDC). Specifically, a negative correlation was detected between DDC mRNA and SARS-CoV-2 RNA levels in in vitro infected epithelial cells and the nasopharyngeal tissue of COVID-19 patients with mild/no symptoms. However, DDC, among other genes related to both DDC expression and SARS-CoV-2-infection (ACE2, dACE2, EPO), was upregulated in these patients, possibly attributed to an orchestrated host antiviral response. Herein, by comparing DDC expression in the nasopharyngeal swab samples of severe/critical to mild COVID-19 cases, we showed a 20 mean-fold reduction, highlighting the importance of the expression of this gene as a potential marker of COVID-19 severity. Moreover, we identified an association of SARS-CoV-2 infection with the expression of key catecholamine biosynthesis/metabolism-related genes, in whole blood samples from hospitalized patients and in cultured cells. Specifically, viral infection downregulated the biosynthetic part of the dopamine pathway (reduction in DDC expression up to 7.5 mean-fold), while enhanced the catabolizing part (increase in monoamine oxidases A and B expression up to 15 and 10 mean-fold, respectively) in vivo, irrespectively of the presence of comorbidities. In accordance, dopamine levels in the sera of severe cases were reduced (up to 3.8 mean-fold). Additionally, a moderate positive correlation between DDC and MAOA mRNA levels (r = 0.527, p < 00001) in the blood was identified upon SARS-CoV-2-infection. These observations were consistent to the gene expression data from SARS-CoV-2-infected Vero E6 and A549 epithelial cells. Furthermore, L-Dopa or dopamine treatment of infected cells attenuated the virus-derived cytopathic effect by 55% and 59%, respectively. The SARS-CoV-2 mediated suppression of dopamine biosynthesis in cell culture was, at least in part, attributed to hypoxia-like conditions triggered by viral infection. These findings suggest that L-Dopa/dopamine intake may have a preventive or therapeutic value for COVID-19 patients.
Collapse
|
31
|
Eroume À Egom E, Shiwani HA, Nouthe B. From acute SARS-CoV-2 infection to pulmonary hypertension. Front Physiol 2022; 13:1023758. [PMID: 36601347 PMCID: PMC9806360 DOI: 10.3389/fphys.2022.1023758] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022] Open
Abstract
As the world progressively recovers from the acute stages of the coronavirus disease 2019 (COVID-19) pandemic, we may be facing new challenges regarding the long-term consequences of COVID-19. Accumulating evidence suggests that pulmonary vascular thickening may be specifically associated with COVID-19, implying a potential tropism of severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) virus for the pulmonary vasculature. Genetic alterations that may influence the severity of COVID-19 are similar to genetic drivers of pulmonary arterial hypertension. The pathobiology of the COVID-19-induced pulmonary vasculopathy shares many features (such as medial hypertrophy and smooth muscle cell proliferation) with that of pulmonary arterial hypertension. In addition, the presence of microthrombi in the lung vessels of individuals with COVID-19 during the acute phase, may predispose these subjects to the development of chronic thromboembolic pulmonary hypertension. These similarities raise the intriguing question of whether pulmonary hypertension (PH) may be a long-term sequela of SARS-COV-2 infection. Accumulating evidence indeed support the notion that SARS-COV-2 infection is indeed a risk factor for persistent pulmonary vascular defects and subsequent PH development, and this could become a major public health issue in the future given the large number of individuals infected by SARS-COV-2 worldwide. Long-term studies assessing the risk of developing chronic pulmonary vascular lesions following COVID-19 infection is of great interest for both basic and clinical research and may inform on the best long-term management of survivors.
Collapse
Affiliation(s)
- Emmanuel Eroume À Egom
- Institut du Savoir Montfort (ISM), University of Ottawa, Ottawa, ON, Canada,CIEL, Centre d’Innovation et de Commercialisation en Recherche Clinique et Bio-Médicale Immânow’EL, Béatitude/Nkolbisson, Yaoundé, Cameroon,Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon,*Correspondence: Emmanuel Eroume À Egom,
| | - Haaris A. Shiwani
- Burnley General Hospital, East Lancashire Hospitals NHS Trust, Burnley, United Kingdom
| | - Brice Nouthe
- Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
32
|
Zuo X, Ren S, Zhang H, Tian J, Tian R, Han B, Liu H, Dong Q, Wang Z, Cui Y, Niu R, Zhang F. Chemotherapy induces ACE2 expression in breast cancer via the ROS-AKT-HIF-1α signaling pathway: a potential prognostic marker for breast cancer patients receiving chemotherapy. J Transl Med 2022; 20:509. [PMID: 36335375 PMCID: PMC9636712 DOI: 10.1186/s12967-022-03716-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Background Angiotensin-converting enzyme 2 (ACE2) is a key enzyme of the renin-angiotensin system and a well-known functional receptor for the entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into host cells. The COVID-19 pandemic has brought ACE2 into the spotlight, and ACE2 expression in tumors and its relationship with SARS-COV-2 infection and prognosis of cancer patients have received extensive attention. However, the association between ACE2 expression and tumor therapy and prognosis, especially in breast cancer, remains ambiguous and requires further investigation. We have previously reported that ACE2 is elevated in drug-resistant breast cancer cells, but the exact function of ACE2 in drug resistance and progression of this malignant disease has not been explored. Methods The expression of ACE2 and HIF-1α in parental and drug-resistant breast cancer cells under normoxic and hypoxic conditions was analyzed by Western blot and qRT-PCR methods. The protein levels of ACE2 in plasma samples from breast cancer patients were examined by ELISA. The relationship between ACE2 expression and breast cancer treatment and prognosis was analyzed using clinical specimens and public databases. The reactive oxygen species (ROS) levels in breast cancer cells were measured by using a fluorescent probe. Small interfering RNAs (siRNAs) or lentivirus-mediated shRNA was used to silence ACE2 and HIF-1α expression in cellular models. The effect of ACE2 knockdown on drug resistance in breast cancer was determined by Cell Counting Kit 8 (CCK-8)-based assay, colony formation assay, apoptosis and EdU assay. Results ACE2 expression is relatively low in breast cancer cells, but increases rapidly and specifically after exposure to anticancer drugs, and remains high after resistance is acquired. Mechanistically, chemotherapeutic agents increase ACE2 expression in breast cancer cells by inducing intracellular ROS production, and increased ROS levels enhance AKT phosphorylation and subsequently increase HIF-1α expression, which in turn upregulates ACE2 expression. Although ACE2 levels in plasma and cancer tissues are lower in breast cancer patients compared with healthy controls, elevated ACE2 in patients after chemotherapy is a predictor of poor treatment response and an unfavorable prognostic factor for survival in breast cancer patients. Conclusion ACE2 is a gene in breast cancer cells that responds rapidly to chemotherapeutic agents through the ROS-AKT-HIF-1α axis. Elevated ACE2 modulates the sensitivity of breast cancer cells to anticancer drugs by optimizing the balance of intracellular ROS. Moreover, increased ACE2 is not only a predictor of poor response to chemotherapy, but is also associated with a worse prognosis in breast cancer patients. Thus, our findings provide novel insights into the spatiotemporal differences in the function of ACE2 in the initiation and progression of breast cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03716-w.
Collapse
|
33
|
Miura Y, Ohkubo H, Nakano A, Bourke JE, Kanazawa S. Pathophysiological conditions induced by SARS-CoV-2 infection reduce ACE2 expression in the lung. Front Immunol 2022; 13:1028613. [PMID: 36405683 PMCID: PMC9673245 DOI: 10.3389/fimmu.2022.1028613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/14/2022] [Indexed: 04/03/2024] Open
Abstract
SARS-CoV-2 infection causes a variety of physiological responses in the lung, and understanding how the expression of SARS-CoV-2 receptor, angiotensin-converting enzyme 2 (ACE2), and its proteolytic activator, transmembrane serine protease 2 (TMPRSS2), are affected in patients with underlying disease such as interstitial pneumonia will be important in considering COVID-19 progression. We examined the expression of ACE2 and TMPRSS2 in an induced usual interstitial pneumonia (iUIP) mouse model and patients with IPF as well as the changes in whole-lung ACE2 and TMPRSS2 expression under physiological conditions caused by viral infection. Histopathological and biochemical characteristics were analyzed using human specimens from patients with IPF and precision-cut lung slices (PCLS) from iUIP mouse model showing UIP with honeycombing and severe fibrosis after non-specific interstitial pneumonia. ACE2 expression decreased with acute lung inflammation and increased in the abnormal lung epithelium of the iUIP mouse model. ACE2 is also expressed in metaplastic epithelial cells. Poly(I:C), interferons, and cytokines associated with fibrosis decreased ACE2 expression in PCLS in the iUIP model. Hypoxia also decreases ACE2 via HIF1α in PCLS. Antifibrotic agent, nintedanib attenuates ACE2 expression in invasive epithelial cells. Patients with IPF are at a higher risk of SARS-CoV-2 infection due to the high expression of ACE2. However, ACE2 and TMPRSS2 expression is decreased by immune intermediaries, including interferons and cytokines that are associated with viral infection and upon administration of antifibrotic agents, suggesting that most of the viral infection-induced pathophysiological responses aid the development of resistance against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yoko Miura
- Department of Neurodevelopmental Disorder Genetics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hirotsugu Ohkubo
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akiko Nakano
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Jane E. Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VA, Australia
| | - Satoshi Kanazawa
- Department of Neurodevelopmental Disorder Genetics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
34
|
Influence of weather factors on the incidence of COVID-19 in Spain. MEDICINA CLÍNICA (ENGLISH EDITION) 2022; 159:255-261. [PMID: 36060101 PMCID: PMC9425111 DOI: 10.1016/j.medcle.2021.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022]
Abstract
Introduction Several studies have analyzed the influence of meteorological and geographical factors on the incidence of COVID-19. Seasonality could be important in the transmission of SARS-CoV-2. This study aims to evaluate the geographical pattern of COVID-19 in Spain and its relationship with different meteorological variables. Methods A provincial ecological study analyzing the influence of meteorological and geographical factors on the cumulative incidence of COVID-19 in the 52 (24 coastal and 28 inland) Spanish provinces during the first three waves was carried out. The cumulative incidence was calculated with data from the National Statistical Institute (INE) and the National Epidemiological Surveillance Network (RENAVE), while the meteorological variables were obtained from the Spanish Meteorological Agency (AEMET). Results The total cumulative incidence, in all three waves, was lower in the coastal provinces than in the inland ones (566 ± 181 vs. 782 ± 154; P = 2.5 × 10−5). The cumulative incidence correlated negatively with mean air temperature (r = −0.49; P = 2.2 × 10−4) and rainfall (r = −0.33; P = .01), and positively with altitude (r = 0.56; P = 1.4 × 10−5). The Spanish provinces with an average temperature <10 °C had almost twice the cumulative incidence than the provinces with temperatures >16 °C. The mean air temperature and rainfall were associated with the cumulative incidence of COVID-19, regardless of other factors (Beta Coefficient of −0.62; P = 3.7 × 10−7 and −0.47; P = 4.2 × 10−5 respectively) Conclusions Meteorological and geographical factors could influence the evolution of the pandemic in Spain. Knowledge regarding the seasonality of the virus would help to predict new waves of COVID-19 infections
Collapse
|
35
|
Piorecka K, Kurjata J, Stanczyk WA. Acriflavine, an Acridine Derivative for Biomedical Application: Current State of the Art. J Med Chem 2022; 65:11415-11432. [PMID: 36018000 PMCID: PMC9469206 DOI: 10.1021/acs.jmedchem.2c00573] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Acriflavine (ACF) has been known for years as an antibacterial
drug. The identification of key oncogenic mechanisms has brought,
in recent years, a significant increase in studies on ACF as a multipurpose
drug that would improve the prognosis for cancer patients. ACF interferes
with the expression of the hypoxia inducible factor, thus acting on
metastatic niches of tumors and significantly enhancing the effects
of other anticancer therapies. It has been recognized as the most
potent HIF-1 inhibitor out of the 336 drugs approved by the FDA. This
work presents up-to-date knowledge about the mechanisms of action
of ACF and its related prodrug systems in the context of anticancer
and SARS-CoV-2 inhibitory properties. It explains the multitask nature
of this drug and suggests mechanisms of ACF’s action on the
coronavirus. Other recent reports on ACF-based systems as potential
antibacterial and antiviral drugs are also described.
Collapse
Affiliation(s)
- Kinga Piorecka
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| | - Jan Kurjata
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| | - Wlodzimierz A Stanczyk
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| |
Collapse
|
36
|
Devaux CA, Raoult D. The impact of COVID-19 on populations living at high altitude: Role of hypoxia-inducible factors (HIFs) signaling pathway in SARS-CoV-2 infection and replication. Front Physiol 2022; 13:960308. [PMID: 36091390 PMCID: PMC9454615 DOI: 10.3389/fphys.2022.960308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Cases of coronavirus disease 2019 (COVID-19) have been reported worldwide. However, one epidemiological report has claimed a lower incidence of the disease in people living at high altitude (>2,500 m), proposing the hypothesis that adaptation to hypoxia may prove to be advantageous with respect to SARS-CoV-2 infection. This publication was initially greeted with skepticism, because social, genetic, or environmental parametric variables could underlie a difference in susceptibility to the virus for people living in chronic hypobaric hypoxia atmospheres. Moreover, in some patients positive for SARS-CoV-2, early post-infection ‘happy hypoxia” requires immediate ventilation, since it is associated with poor clinical outcome. If, however, we accept to consider the hypothesis according to which the adaptation to hypoxia may prove to be advantageous with respect to SARS-CoV-2 infection, identification of the molecular rational behind it is needed. Among several possibilities, HIF-1 regulation appears to be a molecular hub from which different signaling pathways linking hypoxia and COVID-19 are controlled. Interestingly, HIF-1α was reported to inhibit the infection of lung cells by SARS-CoV-2 by reducing ACE2 viral receptor expression. Moreover, an association of the rs11549465 variant of HIF-1α with COVID-19 susceptibility was recently discovered. Here, we review the evidence for a link between HIF-1α, ACE2 and AT1R expression, and the incidence/severity of COVID-19. We highlight the central role played by the HIF-1α signaling pathway in the pathophysiology of COVID-19.
Collapse
Affiliation(s)
- Christian Albert Devaux
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
- Centre National de la Recherche Scientifique, Marseille, France
- *Correspondence: Christian Albert Devaux,
| | - Didier Raoult
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
37
|
Kjertakov M. Commentary: Moderate exercise may prevent the development of severe forms of COVID-19, whereas high-intensity exercise may result in the opposite. Front Physiol 2022; 13:902739. [PMID: 36072850 PMCID: PMC9441653 DOI: 10.3389/fphys.2022.902739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
|
38
|
Amino acid sensor GCN2 promotes SARS-CoV-2 receptor ACE2 expression in response to amino acid deprivation. Commun Biol 2022; 5:651. [PMID: 35778545 PMCID: PMC9249868 DOI: 10.1038/s42003-022-03609-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 06/21/2022] [Indexed: 12/14/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been identified as a primary receptor for severe acute respiratory syndrome coronaviruses 2 (SARS-CoV-2). Here, we investigated the expression regulation of ACE2 in enterocytes under amino acid deprivation conditions. In this study, we found that ACE2 expression was upregulated upon all or single essential amino acid deprivation in human colonic epithelial CCD841 cells. Furthermore, we found that knockdown of general control nonderepressible 2 (GCN2) reduced intestinal ACE2 mRNA and protein levels in vitro and in vivo. Consistently, we revealed two GCN2 inhibitors, GCN2iB and GCN2-IN-1, downregulated ACE2 protein expression in CCD841 cells. Moreover, we found that increased ACE2 expression in response to leucine deprivation was GCN2 dependent. Through RNA-sequencing analysis, we identified two transcription factors, MAFB and MAFF, positively regulated ACE2 expression under leucine deprivation in CCD841 cells. These findings demonstrate that amino acid deficiency increases ACE2 expression and thereby likely aggravates intestinal SARS-CoV-2 infection. Amino acid deprivation increases ACE2 expression in the gut, potentially aggravating SARS-CoV-2 infection.
Collapse
|
39
|
Pouremamali A, Babaei A, Malekshahi SS, Abbasi A, Rafiee N. Understanding the pivotal roles of ACE2 in SARS-CoV-2 infection: from structure/function to therapeutic implication. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022; 23:103. [PMID: 37521846 PMCID: PMC9206724 DOI: 10.1186/s43042-022-00314-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/12/2022] [Indexed: 01/08/2023] Open
Abstract
In December 2019, a novel respiratory tract infection, from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was detected in China that rapidly spread around the world. This virus possesses spike (S) glycoproteins on the surface of mature virions, like other members of coronaviridae. The S glycoprotein is a crucial viral protein for binding, fusion, and entry into the target cells. Binding the receptor-binding domain (RBD) of S protein to angiotensin-converting enzyme 2 (ACE 2), a cell-surface receptor, mediates virus entry into cells; thus, understanding the basics of ACE2 and S protein, their interactions, and ACE2 targeting could be a potent priority for inhibition of virus infection. This review presents current knowledge of the SARS-CoV-2 basics and entry mechanism, structure and organ distribution of ACE2, and also its function in SARS-CoV-2 entry and pathogenesis. Furthermore, it highlights ACE2 targeting by recombinant ACE2 (rACE2), ACE2 activators, ACE inhibitor, and angiotensin II (Ang II) receptor blocker to control the SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Amir Pouremamali
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abouzar Babaei
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Ardeshir Abbasi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nastaran Rafiee
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
40
|
Torices S, Motta CS, da Rosa BG, Marcos AC, Alvarez-Rosa L, Siqueira M, Moreno-Rodriguez T, Matos A, Caetano B, Martins J, Gladulich L, Loiola E, Bagshaw ORM, Stuart JA, Siqueira MM, Stipursky J, Toborek M, Adesse D. SARS-CoV-2 infection of human brain microvascular endothelial cells leads to inflammatory activation through NF-κB non-canonical pathway and mitochondrial remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.06.16.496324. [PMID: 35734080 PMCID: PMC9216721 DOI: 10.1101/2022.06.16.496324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Neurological effects of COVID-19 and long-COVID-19 as well as neuroinvasion by SARS-CoV-2 still pose several questions and are of both clinical and scientific relevance. We described the cellular and molecular effects of the human brain microvascular endothelial cells (HBMECs) in vitro infection by SARS-CoV-2 to understand the underlying mechanisms of viral transmigration through the Blood-Brain Barrier. Despite the low to non-productive viral replication, SARS-CoV-2-infected cultures displayed increased apoptotic cell death and tight junction protein expression and immunolocalization. Transcriptomic profiling of infected cultures revealed endothelial activation via NF-κB non-canonical pathway, including RELB overexpression, and mitochondrial dysfunction. Additionally, SARS-CoV-2 led to altered secretion of key angiogenic factors and to significant changes in mitochondrial dynamics, with increased mitofusin-2 expression and increased mitochondrial networks. Endothelial activation and remodeling can further contribute to neuroinflammatory processes and lead to further BBB permeability in COVID-19.
Collapse
Affiliation(s)
- Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Carolline Soares Motta
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Barbara Gomes da Rosa
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Anne Caroline Marcos
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Liandra Alvarez-Rosa
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, UFRJ, Rio de Janeiro, Brazil
| | - Michele Siqueira
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, UFRJ, Rio de Janeiro, Brazil
| | - Thaidy Moreno-Rodriguez
- Urology Department, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Aline Matos
- Laboratório de Virus Respiratórios e Sarampo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Braulia Caetano
- Laboratório de Virus Respiratórios e Sarampo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Jessica Martins
- Laboratório de Virus Respiratórios e Sarampo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Luis Gladulich
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Erick Loiola
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Olivia RM Bagshaw
- Faculty of Mathematics & Science, Brock University, St. Catharines, Ontario, Canada
| | - Jeffrey A. Stuart
- Faculty of Mathematics & Science, Brock University, St. Catharines, Ontario, Canada
| | - Marilda M. Siqueira
- Laboratório de Virus Respiratórios e Sarampo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Joice Stipursky
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, UFRJ, Rio de Janeiro, Brazil
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Daniel Adesse
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
41
|
Rodriguez Lima DR, Pinzón Rondón ÁM, Rubio Ramos C, Pinilla Rojas DI, Niño Orrego MJ, Díaz Quiroz MA, Molano-González N, Ceballos Quintero JE, Arroyo Santos AF, Ruiz Sternberg ÁM. Clinical characteristics and mortality associated with COVID-19 at high altitude: a cohort of 5161 patients in Bogotá, Colombia. Int J Emerg Med 2022; 15:22. [PMID: 35597911 PMCID: PMC9123834 DOI: 10.1186/s12245-022-00426-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 05/08/2022] [Indexed: 01/08/2023] Open
Abstract
Background There are few data on the clinical outcomes of patients with coronavirus disease 2019 (COVID-19) in cities over 1000 m above sea level (masl). Objectives To describe the clinical characteristics and mortality of patients with COVID-19 treated at a high complexity hospital in Bogotá, Colombia, at 2640 masl. Methods This was an observational study of a cohort including 5161 patients with confirmed COVID-19 infection from 19 March 2020 to 30 April 2021. Demographic data, laboratory values, comorbidities, oxygenation indices, and clinical outcomes were collected. Data were compared between survivors and nonsurvivors. An independent predictive model was performed for mortality and invasive mechanical ventilation (IMV) using classification and regression trees (CART). Results The median cohort age was 66 years (interquartile range (IQR) 53–77), with 1305 patients dying (25%) and 3856 surviving (75%). The intensive care unit (ICU) received 1223 patients (24%). Of 898 patients who received IMV, 613 (68%) of them perished. The ratio of partial pressure arterial oxygen (PaO2) to fraction inspired oxygen (FiO2), or the P/F ratio, upon ICU admission was 105 (IQR 77–146) and 137 (IQR 91–199) in the deceased and survivors, respectively. The CART model showed that the need for IMV, age greater than 79 years, ratio of oxygen saturation (SaO2) to FiO2, or the S/F ratio, less than 259, and lactate dehydrogenase (LDH) greater than 617 U/L at admission were associated with a greater probability of death. Conclusion Among more than 5000 patients with COVID-19 treated in our hospital, mortality at hospital discharge was 25%. Older age, low S/F ratio, and high LDH at admission were predictors of mortality.
Collapse
Affiliation(s)
- David Rene Rodriguez Lima
- Critical and Intensive Care Medicine, Hospital Universitario Mayor-Méderi, Bogotá, Colombia. .,Grupo de Investigación Clínica, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia.
| | - Ángela María Pinzón Rondón
- Grupo de Investigación Clínica, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Cristhian Rubio Ramos
- Critical and Intensive Care Medicine, Hospital Universitario Mayor-Méderi, Bogotá, Colombia
| | | | - Maria José Niño Orrego
- Grupo de Investigación Clínica, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Mateo Andrés Díaz Quiroz
- Grupo de Investigación Clínica, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Nicolás Molano-González
- Grupo de Investigación Clínica, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | | | | | - Ángela María Ruiz Sternberg
- Grupo de Investigación Clínica, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
42
|
Jibaja M, Roldan-Vasquez E, Rello J, Shen H, Maldonado N, Grunauer M, Díaz AM, García F, Ramírez V, Sánchez H, Barberán JL, Paredes JP, Cevallos M, Montenegro F, Puertas S, Briones K, Martínez M, Vélez-Páez J, Montalvo-Villagómez M, Herrera L, Garrido S, Sisa I. Effect of High Altitude on the Survival of COVID-19 Patients in Intensive Care Unit: A Cohort Study. J Intensive Care Med 2022; 37:1265-1273. [PMID: 35532089 PMCID: PMC9095997 DOI: 10.1177/08850666221099827] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Purpose: The effect of high altitude ( ≥ 1500 m) and its potential association with mortality by COVID-19 remains controversial. We assessed the effect of high altitude on the survival/discharge of COVID-19 patients requiring intensive care unit (ICU) admission for mechanical ventilation compared to individuals treated at sea level. Methods: A retrospective cohort multi-center study of consecutive adults patients with a positive RT-PCR test for COVID-19 who were mechanically ventilated between March and November 2020. Data were collected from two sea-level hospitals and four high-altitude hospitals in Ecuador. The primary outcome was ICU and hospital survival/discharge. Survival analysis was conducted using semi-parametric Cox proportional hazards models. Results: Of the study population (n = 670), 35.2% were female with a mean age of 58.3 ± 12.6 years. On admission, high-altitude patients were more likely to be younger (57.2 vs. 60.5 years old), presented with less comorbidities such as hypertension (25.9% vs. 54.9% with p-value <.001) and diabetes mellitus (20.5% vs. 37.2% with p-value <.001), less probability of having a capillary refill time > 3 sec (13.7% vs. 30.1%, p-value <.001), and less severity-of-illness condition (APACHE II score, 17.5 ± 8.1 vs. 20 ± 8.2, p < .01). After adjusting for key confounders high altitude is associated with significant higher probabilities of ICU survival/discharge (HR: 1.74 [95% CI: 1.46-2.08]) and hospital survival/discharge (HR: 1.35 [95% CI: 1.18-1.55]) than patients treated at sea level. Conclusions: Patients treated at high altitude at any time point during the study period were 74% more likely to experience ICU survival/discharge and 35% more likely to experience hospital survival/discharge than to the sea-level group. Possible reasons for these findings are genetic and physiological adaptations due to exposure to chronic hypoxia.
Collapse
Affiliation(s)
- Manuel Jibaja
- Escuela de Medicina, Universidad Internacional del Ecuador UIDE, Quito, Ecuador
- Unidad de Cuidados Intensivos, Hospital de Especialidades Eugenio Espejo, Quito, Ecuador
| | - Estefania Roldan-Vasquez
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Jordi Rello
- Vall d’Hebrón Institute of Research (VHIR), Centro de Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
- CHRU Nîmes, Nîmes, France
| | - Hua Shen
- Department of Mathematics and Statistics, University of Calgary, Calgary, Alberta, Canada
| | - Nelson Maldonado
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Michelle Grunauer
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Ana María Díaz
- Unidad de Cuidados Intensivos, Hospital de Especialidades Eugenio Espejo, Quito, Ecuador
| | - Fernanda García
- Unidad de Cuidados Intensivos, Hospital de Especialidades Eugenio Espejo, Quito, Ecuador
| | - Vanessa Ramírez
- Unidad de Cuidados Intensivos, Hospital de Especialidades Eugenio Espejo, Quito, Ecuador
| | - Hernán Sánchez
- Unidad de Cuidados Intensivos, Hospital de Especialidades Eugenio Espejo, Quito, Ecuador
| | - José Luis Barberán
- Unidad de Cuidados Intensivos, Hospital de Especialidades, Portoviejo, Ecuador
| | - Juan Pablo Paredes
- Unidad de Cuidados Intensivos, Hospital de Especialidades, Portoviejo, Ecuador
| | - Mónica Cevallos
- Unidad de Cuidados Intensivos, Hospital de Especialidades, Portoviejo, Ecuador
| | - Francisco Montenegro
- Unidad de Cuidados Intensivos, Hospital de Especialidades José Carrasco Arteaga, Cuenca, Ecuador
| | - Soraya Puertas
- Unidad de Cuidados Intensivos, Hospital de Especialidades José Carrasco Arteaga, Cuenca, Ecuador
| | - Killen Briones
- Unidad de Cuidados Intensivos, Hospital del IESS, Babahoyo, Ecuador
| | - Marlon Martínez
- Unidad de Cuidados Intensivos, Hospital del IESS, Babahoyo, Ecuador
| | - Jorge Vélez-Páez
- Unidad de Cuidados Intensivos, Hospital General Pablo Arturo Suárez, Quito, Ecuador
| | | | - Luis Herrera
- Unidad de Cuidados Intensivos, Hospital del IESS, Ibarra, Ecuador
| | - Santiago Garrido
- Unidad de Cuidados Intensivos, Hospital del IESS, Ibarra, Ecuador
| | - Ivan Sisa
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| |
Collapse
|
43
|
Shukla P, Chopada K, Sakure A, Hati S. Current Trends and Applications of Food-derived Antihypertensive
Peptides for the Management of Cardiovascular Disease. Protein Pept Lett 2022; 29:408-428. [DOI: 10.2174/0929866529666220106100225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/26/2021] [Accepted: 11/20/2021] [Indexed: 11/22/2022]
Abstract
Abstract:
Food derived Antihypertensive peptides is considered as a natural supplement for controlling the hypertension. Food protein not only serve as a macronutrient but also act as raw material for biosynthesis of physiologically active peptides. Food sources like milk and milk products, animal protein such as meat, chicken, fish, eggs and plant derived proteins from soy, rice, wheat, mushroom, pumpkins contain high amount of antihypertensive peptides. The food derived antihypertensive peptides has ability to supress the action of rennin and Angiotesin converting enzyme (ACE) which is mainly involved in regulation of blood pressure by RAS. The biosynthesis of endothelial nitric oxide synthase is also improved by ACE inhibitory peptides which increase the production of nitric oxide in vascular walls and encourage vasodilation. Interaction between the angiotensin II and its receptor is also inhibited by the peptides which help to reduce hypertension. This review will explore the novel sources and applications of food derived peptides for the management of hypertension.
Collapse
Affiliation(s)
- Pratik Shukla
- Department of Dairy Microbiology, SMC College of Dairy Science, Anand Agricultural University, Anand- 388110,
Gujarat, India
| | - Keval Chopada
- Department of Dairy Microbiology, SMC College of Dairy Science, Anand Agricultural University, Anand- 388110,
Gujarat, India
| | - Amar Sakure
- Department of Agricultural Biotechnology, Anand Agricultural University, Anand- 388110, Gujarat,
India
| | - Subrota Hati
- Department of Dairy Microbiology, SMC College of Dairy Science, Anand Agricultural University, Anand- 388110,
Gujarat, India
| |
Collapse
|
44
|
Halawa S, Pullamsetti SS, Bangham CRM, Stenmark KR, Dorfmüller P, Frid MG, Butrous G, Morrell NW, de Jesus Perez VA, Stuart DI, O'Gallagher K, Shah AM, Aguib Y, Yacoub MH. Potential long-term effects of SARS-CoV-2 infection on the pulmonary vasculature: a global perspective. Nat Rev Cardiol 2022; 19:314-331. [PMID: 34873286 PMCID: PMC8647069 DOI: 10.1038/s41569-021-00640-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 12/13/2022]
Abstract
The lungs are the primary target of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, with severe hypoxia being the cause of death in the most critical cases. Coronavirus disease 2019 (COVID-19) is extremely heterogeneous in terms of severity, clinical phenotype and, importantly, global distribution. Although the majority of affected patients recover from the acute infection, many continue to suffer from late sequelae affecting various organs, including the lungs. The role of the pulmonary vascular system during the acute and chronic stages of COVID-19 has not been adequately studied. A thorough understanding of the origins and dynamic behaviour of the SARS-CoV-2 virus and the potential causes of heterogeneity in COVID-19 is essential for anticipating and treating the disease, in both the acute and the chronic stages, including the development of chronic pulmonary hypertension. Both COVID-19 and chronic pulmonary hypertension have assumed global dimensions, with potential complex interactions. In this Review, we present an update on the origins and behaviour of the SARS-CoV-2 virus and discuss the potential causes of the heterogeneity of COVID-19. In addition, we summarize the pathobiology of COVID-19, with an emphasis on the role of the pulmonary vasculature, both in the acute stage and in terms of the potential for developing chronic pulmonary hypertension. We hope that the information presented in this Review will help in the development of strategies for the prevention and treatment of the continuing COVID-19 pandemic.
Collapse
Affiliation(s)
| | - Soni S Pullamsetti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University, Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Charles R M Bangham
- Department of Infectious Diseases, Faculty of Medicine, Imperial College London, London, UK
| | - Kurt R Stenmark
- Divisions of Paediatric Critical Care Medicine and Cardiovascular Pulmonary Research, University of Colorado Denver, Denver, CO, USA
| | - Peter Dorfmüller
- Department of Pathology, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University, Giessen, Germany
| | - Maria G Frid
- Divisions of Paediatric Critical Care Medicine and Cardiovascular Pulmonary Research, University of Colorado Denver, Denver, CO, USA
| | - Ghazwan Butrous
- Medway School of Pharmacy, University of Kent at Canterbury, Canterbury, UK
| | - Nick W Morrell
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Vinicio A de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - David I Stuart
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Kevin O'Gallagher
- King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Yasmine Aguib
- Aswan Heart Centre, Aswan, Egypt
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Magdi H Yacoub
- Aswan Heart Centre, Aswan, Egypt.
- National Heart & Lung Institute, Imperial College London, London, UK.
- Harefield Heart Science Centre, London, UK.
| |
Collapse
|
45
|
Zhang Q, Ling S, Hu K, Liu J, Xu JW. Role of the renin-angiotensin system in NETosis in the coronavirus disease 2019 (COVID-19). Pharmacotherapy 2022; 148:112718. [PMID: 35176710 PMCID: PMC8841219 DOI: 10.1016/j.biopha.2022.112718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/20/2022]
Abstract
Myocardial infarction and stroke are the leading causes of death in the world. Numerous evidence has confirmed that hypertension promotes thrombosis and induces myocardial infarction and stroke. Recent findings reveal that neutrophil extracellular traps (NETs) are involved in the induction of myocardial infarction and stroke. Meanwhile, patients with severe COVID-19 suffer from complications such as myocardial infarction and stroke with pathological signs of NETs. Due to the extremely low amount of virus detected in the blood and remote organs (e.g., heart, brain and kidney) in a few cases, it is difficult to explain the mechanism by which the virus triggers NETosis, and there may be a different mechanism than in the lung. A large number of studies have found that the renin-angiotensin system regulates the NETosis at multiple levels in patients with COVID-19, such as endocytosis of SARS-COV-2, abnormal angiotensin II levels, neutrophil activation and procoagulant function at multiple levels, which may contribute to the formation of reticular structure and thrombosis. The treatment of angiotensin-converting enzyme inhibitors (ACEI), angiotensin II type 1 receptor blockers (ARBs) and neutrophil recruitment and active antagonists helps to regulate blood pressure and reduce the risk of net and thrombosis. The review will explore the possible role of the angiotensin system in the formation of NETs in severe COVID-19.
Collapse
|
46
|
Hu J, Han Z, Heidari AA, Shou Y, Ye H, Wang L, Huang X, Chen H, Chen Y, Wu P. Detection of COVID-19 severity using blood gas analysis parameters and Harris hawks optimized extreme learning machine. Comput Biol Med 2022; 142:105166. [PMID: 35077935 PMCID: PMC8701842 DOI: 10.1016/j.compbiomed.2021.105166] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023]
Abstract
Coronavirus disease-2019 (COVID-19) has made the world more cautious about widespread viruses, and a tragic pandemic that was caused by a novel coronavirus has harmed human beings in recent years. The new coronavirus pneumonia outbreak is spreading rapidly worldwide. We collect arterial blood samples from 51 patients with a COVID-19 diagnosis. Blood gas analysis is performed using a Siemens RAPID Point 500 blood gas analyzer. To accurately determine the factors that play a decisive role in the early recognition and discrimination of COVID-19 severity, a prediction framework that is based on an improved binary Harris hawk optimization (HHO) algorithm in combination with a kernel extreme learning machine is proposed in this paper. This method uses specular reflection learning to improve the original HHO algorithm and is referred to as HHOSRL. The experimental results show that the selected indicators, such as age, partial pressure of oxygen, oxygen saturation, sodium ion concentration, and lactic acid, are essential for the early accurate assessment of COVID-19 severity by the proposed feature selection method. The simulation results show that the established methodlogy can achieve promising performance. We believe that our proposed model provides an effective strategy for accurate early assessment of COVID-19 and distinguishing disease severity. The codes of HHO will be updated in https://aliasgharheidari.com/HHO.html.
Collapse
Affiliation(s)
- Jiao Hu
- Department of Computer Science and Artificial Intelligence, Wenzhou University, Wenzhou, 325035, China.
| | - Zhengyuan Han
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China.
| | - Ali Asghar Heidari
- School of Surveying and Geospatial Engineering, College of Engineering, University of Tehran, Tehran, Iran.
| | - Yeqi Shou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China.
| | - Hua Ye
- Department of Pulmonary and Critical Care Medicine, Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing, 325600, China.
| | - Liangxing Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China.
| | - Xiaoying Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China.
| | - Huiling Chen
- Department of Computer Science and Artificial Intelligence, Wenzhou University, Wenzhou, 325035, China.
| | - Yanfan Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China.
| | - Peiliang Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China.
| |
Collapse
|
47
|
Song Y, Jia H, Hua Y, Wu C, Li S, Li K, Liang Z, Wang Y. The Molecular Mechanism of Aerobic Exercise Improving Vascular Remodeling in Hypertension. Front Physiol 2022; 13:792292. [PMID: 35295586 PMCID: PMC8919036 DOI: 10.3389/fphys.2022.792292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 01/13/2022] [Indexed: 11/26/2022] Open
Abstract
The treatment and prevention of hypertension has been a worldwide medical challenge. The key pathological hallmark of hypertension is altered arterial vascular structure and function, i.e., increased peripheral vascular resistance due to vascular remodeling. The aim of this review is to elucidate the molecular mechanisms of vascular remodeling in hypertension and the protective mechanisms of aerobic exercise against vascular remodeling during the pathological process of hypertension. The main focus is on the mechanisms of oxidative stress and inflammation in the pathological condition of hypertension and vascular phenotypic transformation induced by the trilaminar structure of vascular endothelial cells, smooth muscle cells and extracellular matrix, and the peripheral adipose layer of the vasculature. To further explore the possible mechanisms by which aerobic exercise ameliorates vascular remodeling in the pathological process of hypertension through anti-proliferative, anti-inflammatory, antioxidant and thus inhibiting vascular phenotypic transformation. It provides a new perspective to reveal the intervention targets of vascular remodeling for the prevention and treatment of hypertension and its complications.
Collapse
Affiliation(s)
- Yinping Song
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi’an, China
| | - Hao Jia
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi’an, China
| | - Yijie Hua
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi’an, China
| | - Chen Wu
- School of Health and Sports, Xi’an Fanyi University, Xi’an, China
| | - Sujuan Li
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi’an, China
| | - Kunzhe Li
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi’an, China
| | - Zhicheng Liang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi’an, China
| | - Youhua Wang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi’an, China
- *Correspondence: Youhua Wang,
| |
Collapse
|
48
|
Cajanding RJM. Silent Hypoxia in COVID-19 Pneumonia: State of Knowledge, Pathophysiology, Mechanisms, and Management. AACN Adv Crit Care 2022; 33:143-153. [PMID: 35113990 DOI: 10.4037/aacnacc2022448] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Patients with COVID-19 often present with life-threatening hypoxemia without dyspnea or signs of respiratory distress. Termed silent or happy hypoxia, it has puzzled clinicians and challenged and defied our understanding of normal respiratory physiology. A range of host- and pathogen-related factors appears to contribute to its development, including SARS-CoV-2's ability to produce different COVID-19 phenotypes; induce endothelial damage and elicit a vascular distress response; invade cells of the central nervous system and disrupt normal interoception and response; and modulate transcription factors involved in hypoxic responses. Because hypoxemia in COVID-19 is associated with increased mortality risk and poorer survival, early detection and prompt treatment is essential to prevent potential complications. Interventions to prevent hypoxemia and improve oxygen delivery to the blood and the tissues include home pulse-oximetry monitoring, optimization of patient positioning, judicious use of supplemental oxygen, breathing control exercises, and timely and appropriate use of ventilatory modalities and adjuncts.
Collapse
Affiliation(s)
- Ruff Joseph Macale Cajanding
- Ruff Joseph Macale Cajanding is Senior Charge Nurse, Adult Critical Care Unit, 6th Floor, King George V Building, St Bartholomew's Hospital, Barts Health NHS Trust, 2 King Edward Street, London EC1A 1HQ, United Kingdom
| |
Collapse
|
49
|
Emmi A, Boura I, Raeder V, Mathew D, Sulzer D, Goldman JE, Leta V. Covid-19, nervous system pathology, and Parkinson's disease: Bench to bedside. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 165:17-34. [PMID: 36208899 PMCID: PMC9361071 DOI: 10.1016/bs.irn.2022.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Coronavirus disease 2019 (Covid-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection is primarily regarded as a respiratory disease; however, multisystemic involvement accompanied by a variety of clinical manifestations, including neurological symptoms, are commonly observed. There is, however, little evidence supporting SARS-CoV-2 infection of central nervous system cells, and neurological symptoms for the most part appear to be due to damage mediated by hypoxic/ischemic and/or inflammatory insults. In this chapter, we report evidence on candidate neuropathological mechanisms underlying neurological manifestations in Covid-19, suggesting that while there is mostly evidence against SARS-CoV-2 entry into brain parenchymal cells as a mechanism that may trigger Parkinson's disease and parkinsonism, that there are multiple means by which the virus may cause neurological symptoms.
Collapse
Affiliation(s)
- Aron Emmi
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padova, Italy
| | - Iro Boura
- Department of Neurology, University Hospital of Heraklion, Crete, Greece
| | - Vanessa Raeder
- Parkinson's Foundation Centre of Excellence, King's College Hospital NHS Foundation Trust, London, United Kingdom; Department of Neurology, Technical University Dresden, Dresden, Germany; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Donna Mathew
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - David Sulzer
- Departments of Psychiatry, Neurology, Pharmacology, Columbia University Medical Center, New York State Psychiatric Institute, New York, United States
| | - James E Goldman
- Department of Pathology and Cell Biology, and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, New York, NY, United States
| | - Valentina Leta
- Parkinson's Foundation Centre of Excellence, King's College Hospital NHS Foundation Trust, London, United Kingdom; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
50
|
Mahdavinezhad F, Farmani AR, Pakniat H, Taghavi S, Gharaei R, Valipour J, Amidi F. COVID-19 and varicocele: the possible overlap factors and the common therapeutic approaches. Am J Reprod Immunol 2021; 87:e13518. [PMID: 34967487 DOI: 10.1111/aji.13518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/02/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
Varicocele is recognized as one of the main attributable causes of male infertility which can affect spermatogenesis by various pathophysiological mechanisms. Recent studies have identified oxidative stress and reduction in antioxidant, hyperthermia, hypoxia, hormonal dysfunction, and inflammatory conditions as major factors in the pathophysiology of varicocele, all of which have known direct associations with the coronavirus disease 2019 (COVID-19) and can significantly increase the risk of detrimental COVID-19-related outcomes. Emerging data have shown an association between COVID-19 and inflammation, overproduction of cytokine, and other pathophysiological processes. The present review, summarizes the current understanding of the pathophysiology of varicocele and investigates the potential correlation between the severity of COVID-19 and the varicocele disease. In addition, various possible treatments which can be effective in both diseases were examined. Despite numerous challenges associated with the prevalence of COVID-19 in healthcare systems in infected countries, special attention should be given to maintaining a high level of care for complex patients with a pre-existing disease such as varicocele and providing appropriate practical advice for optimal control of the COVID-19 disease. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Forough Mahdavinezhad
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Farmani
- Department of Tissue Engineering, Faculty of Advanced Technologies, Tehran University of Medical Sciences, Tehran, Iran.,Tissue Engineering Department, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Hamideh Pakniat
- Department of Obstetrics and Gynecology, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Saeed Taghavi
- Iran Polymer and Petrochemical Institute, Tehran, Iran
| | - Roghaye Gharaei
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamal Valipour
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fardin Amidi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|