1
|
Huang S, Zhou Y, Ji H, Zhang T, Liu S, Ma L, Deng D, Ding Y, Han L, Shu S, Wang Y, Chen X. Decoding mechanisms and protein markers in lung-brain axis. Respir Res 2025; 26:190. [PMID: 40390067 PMCID: PMC12090670 DOI: 10.1186/s12931-025-03272-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 05/08/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND The lung-brain axis represents a complex bidirectional communication network that is pivotal in the crosstalk between respiratory and neurological functions. This review summarizes the current understanding of the mechanisms and protein markers that mediate the effects of lung diseases on brain health. MAIN FINDINGS In this review, we explore the mechanisms linking lung injury to neurocognitive impairments, focusing on neural pathways, immune regulation and inflammatory responses, microorganism pathways, and hypoxemia. Specifically, we highlight the role of the vagus nerve in modulating the central nervous system response to pulmonary stimuli; Additionally, the regulatory function of the immune system is underscored, with evidence suggesting that lung-derived immune mediators can traverse the blood-brain barrier, induce neuroinflammation and cognitive decline; Furthermore, we discuss the potential of lung microbiota to influence brain diseases through microbial translocation and immune activation; Finally, the impact of hypoxemia is examined, with findings indicating that it can exacerbate cerebral injury via oxidative stress and impaired perfusion. Moreover, we analyze how pulmonary conditions, such as pneumonia, ALI/ARDS, and asthma, contribute to neurological dysfunction. Prolonged mechanical ventilation can also contribute to cognitive impairment. Conversely, brain diseases (e.g., stroke, traumatic brain injury) can lead to acute respiratory complications. In addition, protein markers such as TLR4, ACE2, A-SAA, HMGB1, and TREM2 are crucial to the lung-brain axis and correlate with disease severity. We also discuss emerging therapeutic strategies targeting this axis, including immunomodulation and microbiome engineering. Overall, understanding the lung-brain interplay is crucial for developing integrated treatment strategies and improving patient outcomes. Further research is needed to elucidate the molecular mechanisms and foster interdisciplinary collaboration.
Collapse
Affiliation(s)
- Shiqian Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Yuxi Zhou
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Haipeng Ji
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Shiya Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Lulin Ma
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Daling Deng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Yuanyuan Ding
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Linlin Han
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Shaofang Shu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Yu Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China.
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China.
| |
Collapse
|
2
|
Yang L, Zhou X, Liu J, Yang G, Yu J, Tan W, Fang X, Li W, He J, Ma Q, Yu L, Lu Z. Liang-Ge-San attenuates virus-induced acute lung injury by targeting FXR-mediated ACE2 downregulation to modulate the formation of the cytokine storm. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156584. [PMID: 40056637 DOI: 10.1016/j.phymed.2025.156584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Traditional Chinese medicine has been recognized for its significant role in treating acute lung injury (ALI) due to its distinct therapeutic advantages. Liang-Ge-San (LGS), a formulation from the ancient "Taiping Huimin Hejiju Fang", is believed to possess beneficial effects for treating ALI. However, LGS's precise mechanisms and efficacy in addressing viral ALI remain inadequately explored. PURPOSE To evaluate LGS's therapeutic effects and underlying mechanisms in treating viral-induced ALI. METHODS The protective effects of LGS were examined in a Polyinosinic-polycytidylic acid [Poly(I:C)]-induced ALI model using real-time quantitative PCR, enzyme-linked immunosorbent assay, and histopathological analysis. A bioinformatics approach combined with network pharmacology was utilized to ascertain the key targets of LGS in viral pneumonia. The pharmacodynamic mechanisms of LGS in viral ALI were further validated through immunofluorescence, overexpression, short hairpin RNA, chromatin immunoprecipitation, and target agonist assays. RESULTS LGS administration resulted in a reduction of IL-1β, IL-6, and TNF-α levels, along with a decrease in macrophage infiltration, pulmonary damage, and pneumonedema following the Poly(I:C) challenge. Bioinformatics and network pharmacology analyses suggested that Farnesyl X receptor (FXR) and angiotensin converting enzyme 2 (ACE2) are potential therapeutic targets for LGS in viral pneumonia. Further experiments revealed that LGS suppressed the expression of FXR, ACE2, and NF-κB-p65 in Poly(I:C)-infected cells. Notably, overexpression of FXR counteracted the repressive effects of LGS, while ACE2 expression remained unchanged in FXR-knockdown RAW264.7 cells upon treatment with Poly(I:C) or LGS. Additionally, LGS inhibited the interaction between FXR and ACE2 transcriptional promoters. In vivo, LGS attenuated the Poly(I:C)-induced upregulation of FXR, ACE2, IL-1β, IL-6, and TNF-α in ALI zebrafish and mice models, effects that could be reversed by chenodeoxycholic acid (CDCA), an FXR agonist. Moreover, LGS markedly alleviated weight loss, improved survival rates, reduced lung index, diminished viral load, and inhibited lung pathological changes in H1N1-PR8-induced ALI mice. IL-1β, IL-6, TNF-α, INF-γ, FXR, ACE2, small heterodimer partner, and NF-κB-p65 levels were markedly reduced by LGS, with these effects being reversed by CDCA. CONCLUSION This investigation provides the first evidence that FXR/ACE2 signaling is pivotal in acute respiratory viral infections, while LGS demonstrates antiviral activity against viral-induced ALI. LGS inhibits ACE2 expression induced by viral infection via FXR inhibition and modulates the cytokine storm, thus alleviating viral ALI. These findings suggest that LGS may be a promising treatment strategy for treating viral ALI.
Collapse
Affiliation(s)
- Liling Yang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Xiangjun Zhou
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Junshan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Guangli Yang
- Department of Central Laboratory, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Weifu Tan
- Department of Neonatology, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Xiaochuan Fang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Wei Li
- Department of Neonatology, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Jiayang He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, PR China
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, PR China.
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| | - Zibin Lu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
3
|
Zheng J, Pang Q, Fu Z. Replication Features of SARS-CoV-2 and Advantages of Targeting S Protein with Aptamers to Block Viral Entry. ACS OMEGA 2025; 10:15840-15851. [PMID: 40321580 PMCID: PMC12044446 DOI: 10.1021/acsomega.4c10933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 05/08/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a virus of the coronaviridae family. The virus enters the cell through binding to the corresponding receptor angiotensin-converting enzyme 2 (ACE2) on host cell membrane with the spike protein (S protein) on its envelope; thus, we can design inhibitors that bind the S protein to block the entry of the virus into cells. Aptamers are single stranded DNA or RNA molecules that can form specific three-dimensional structures and bind their target molecules with high affinity and specificity and thus are promising candidates for S protein inhibitors. This paper reviews the replication cycle and cell entry mechanisms of SARS-CoV-2 as well as the preparation principle and characteristics of aptamers, features a discussion of the advantages of using aptamers to target the S protein to prevent SARS-CoV-2 from infecting cells, and finally summarizes the research progress in S protein-blocking aptamers.
Collapse
Affiliation(s)
| | | | - Zhaoying Fu
- Yan’an Medical College, Yan’an University, Yan’an 716000, China
| |
Collapse
|
4
|
Lin MW, Lin CH, Chang JR, Chiang HH, Wu TH, Lin CS. The influence of PM2.5 exposure on SARS-CoV-2 infection via modulating the expression of angiotensin converting enzyme II. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136887. [PMID: 39700942 DOI: 10.1016/j.jhazmat.2024.136887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Particulate matter 2.5 (PM2.5) pollution and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic are the greatest environmental health issues worldwide. Several statistics revealed the significant positive correlation between the morbidity of coronavirus disease-19 (COVID-19) and the levels of air pollution. Nevertheless, there is no direct experimental evidence to indicate the effect of PM2.5 exposure on SARS-CoV-2 infection. The objective of this study was to evaluate whether the infection of SARS-CoV-2 affected by PM2.5 through angiotensin-converting enzyme II (ACE2) expression enhances and investigate the function of ACE2 in lung injury induced by PM2.5. An animal model of PM2.5-induced lung injury was established using wild-type (WT, C57BL/6), human ACE2 transgenic (K18-hACE2 TG), and murine ACE2 gene knockout (mACE2 KO) mice. The results indicate that PM2.5 exposure facilitates SARS-CoV-2 infection through inducing ACE2 expression in vitro (10 μg/mL) and in vivo (6.25 mg/kg/day in 50 μL saline). The levels of ACE, inflammatory cytokines, and mitogen-activated protein kinase (MAPK) proteins in WT, K18-hACE TG and mACE2 KO mice were significantly increased after PM2.5 instillation. The severest PM2.5-induced lung damage was observed in mACE2 KO mice. In summary, ACE2 plays a double-edged sword role in lung injury, PM2.5 exposure contributed to SARS-CoV-2 infection through inducing ACE2 expression, but ACE2 also protected pulmonary inflammation from PM2.5 challenge.
Collapse
Affiliation(s)
- Meng-Wei Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Cheng-Han Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Jia-Rong Chang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Hua-Hsin Chiang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Ting-Hsuan Wu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Chih-Sheng Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| |
Collapse
|
5
|
Islam MA, Ford Versypt AN. Mathematical modeling of impacts of patient differences on renin-angiotensin system and applications to COVID-19 lung fibrosis outcomes. Comput Biol Med 2025; 186:109631. [PMID: 39753028 PMCID: PMC11932320 DOI: 10.1016/j.compbiomed.2024.109631] [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: 08/10/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 02/20/2025]
Abstract
Patient-specific premorbidity, age, and sex are significant heterogeneous factors that influence the severe manifestation of lung diseases, including COVID-19 fibrosis. The renin-angiotensin system (RAS) plays a prominent role in regulating the effects of these factors. Recent evidence shows patient-specific alterations of RAS peptide homeostasis concentrations with premorbidity and the expression level of angiotensin-converting enzyme 2 (ACE2) during COVID-19. However, conflicting evidence suggests decreases, increases, or no changes in RAS peptides after SARS-CoV-2 infection. A multiscale computational model was developed to quantify the systemic contribution of heterogeneous factors of RAS during COVID-19. Three submodels were connected-an agent-based model for in-host COVID-19 response in the lung tissue, a RAS dynamics model, and a fibrosis dynamics model to investigate the effects of patient-group-specific factors in the systemic alteration of RAS and collagen deposition in the lung. The model results indicated cell death due to inflammatory response as a major contributor to the reduction of ACE and ACE2. The model explained possible mechanisms for conflicting evidence of patient-group-specific changes in RAS peptides in previously published studies. RAS peptides decreased for all virtual patient groups with aging in both sexes. In contrast, large variations in the magnitude of reduction were observed between male and female virtual patients in the older and middle-aged groups. The patient-specific variations in homeostasis RAS peptide concentrations of SARS-CoV-2-negative patients affected the dynamics of RAS during infection. This model may find further applications in patient-specific calibrations of tissue models for acute and chronic lung diseases to develop personalized treatments.
Collapse
Affiliation(s)
- Mohammad Aminul Islam
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Ashlee N Ford Versypt
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA; Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA; Institute for Artificial Intelligence and Data Science, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA; Witebsky Center for Microbial Pathogenesis and Immunology, University at Buffalo, The State University of New York, Buffalo, NY, 14203, USA; Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14215, USA.
| |
Collapse
|
6
|
Schaich CL, Chappell MC, Shotwell MS, Joly MM, Gibbs KW, Barksdale A, Douglas IS, Chen P, Levitt JE, Puskarich MA, Rice TW, Harkins MS, Hudock KM, Lanspa MJ, Ginde AA, Self WH, Collins SP, Files DC. The circulating renin-angiotensin system and mortality among patients hospitalized for COVID-19: a mechanistic substudy of the ACTIV-4 Host Tissue trials. Am J Physiol Lung Cell Mol Physiol 2025; 328:L405-L412. [PMID: 39884670 DOI: 10.1152/ajplung.00372.2024] [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: 11/18/2024] [Revised: 12/26/2024] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
SARS-CoV-2 targets angiotensin-converting enzyme-2 (ACE2), a key peptidase of the renin-angiotensin system (RAS), which regulates the balance of the vasoconstrictor/inflammatory peptide Ang II and the vasodilator/anti-inflammatory peptide Ang-(1-7). Few studies have quantified the circulating elements of the RAS longitudinally in SARS-CoV-2 infection and their association with COVID-19 outcomes. Thus, we evaluated the association of circulating RAS enzymes and peptides with mortality among patients with COVID-19. Blood samples were collected from 111 patients with COVID-19 and new-onset hypoxemia during the delta and omicron waves at 19 hospitals in the United States. Circulating RAS components were quantified via radioimmunoassay or ELISA at 0 (baseline), 1, 3, and 5 days after randomization. We used multivariable Cox regression to estimate the association of baseline and longitudinal RAS concentrations with 90-day mortality. Participants were aged 18-90 (means [SD]: 55 [14]) yr and 62% were male. There were 22 (20%) deaths over 90 days of follow-up. ACE2 levels above the sample median (≥4.9 pM; adjusted HR [95% CI]: 0.10 [0.02, 0.43]) and ACE2/ACE ratio (≥6.0 × 10-3; adjusted HR: 0.08 [0.02, 0.39]) were associated with significantly lower mortality. Similarly, when analyzed as continuous, log2-normalized, time-varying predictors from day 0 to day 5, twofold increments of ACE2 and ACE2/ACE ratio over this period were associated with lower mortality (adjusted HR: 0.79 [0.65, 0.97] and 0.78 [0.63, 0.97], respectively). Circulating Ang II, Ang-(1-7), and ACE levels were not associated with mortality. These results suggest higher circulating ACE2 protein in hospitalized patients with COVID-19 is associated with reduced mortality.NEW & NOTEWORTHY We measured circulating components of the renin-angiotensin system (RAS) longitudinally over 5 days among patients hospitalized with COVID-19 and new-onset hypoxemia. We found that higher serum angiotensin-converting enzyme (ACE)-2 protein and ACE2/ACE ratio, both at baseline and when analyzed as time-varying, repeated measures, were associated with lower 90-day mortality. Results suggest a role for circulating ACE2 as a biomarker of adverse outcomes and could inform treatment strategies targeting the RAS in severe COVID-19 illness.
Collapse
Affiliation(s)
- Christopher L Schaich
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Mark C Chappell
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Matthew S Shotwell
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Meghan M Joly
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Kevin W Gibbs
- Department of Internal Medicine, Section of Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Aaron Barksdale
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Ivor S Douglas
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, Denver Health Medical Center, Anschutz School of Medicine, University of Colorado, Denver, Colorado, United States
| | - Peter Chen
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Joseph E Levitt
- Department of Medicine, Stanford University, Stanford, California, United States
| | - Michael A Puskarich
- Department of Emergency Medicine, Hennepin Healthcare, Minneapolis, Minnesota, United States
| | - Todd W Rice
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt Institute for Clinical Trials Research, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Michelle S Harkins
- Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico, United States
| | - Kristin M Hudock
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio, United States
| | - Michael J Lanspa
- Department of Pulmonary/Critical Care Medicine, Intermountain Medical Center, Murray, Utah, United States
| | - Adit A Ginde
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Wesley H Self
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Sean P Collins
- Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Geriatric Research and Education Center, Veterans Affairs Tennessee Valley Health System, Nashville, Tennessee, United States
| | - D Clark Files
- Department of Internal Medicine, Section of Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| |
Collapse
|
7
|
Li R, Zhang J, Ren L. A Meta-Analysis of the Impact of Using Angiotensin-Converting Enzyme Inhibitors (ACEIs) or Angiotensin II Receptor Blockers (ARBs) on Mortality, Severity, and Healthcare Resource Utilization in Patients with COVID-19. Adv Respir Med 2025; 93:4. [PMID: 39996621 PMCID: PMC11852372 DOI: 10.3390/arm93010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 01/26/2025] [Indexed: 02/26/2025]
Abstract
OBJECTIVE The primary objective of this study is to explore the potential link between the utilization of angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin II receptor blockers (ARBs) and its impact on mortality, disease severity, and healthcare resource utilization in individuals diagnosed with COVID-19. We aim to establish a solid theoretical foundation for safe and effective clinical medications. METHODS We conducted a comprehensive search of various databases, including CNKI, PubMed, Science, Cell, Springer, Nature, Web of Science, and Embase. We also traced the literature of the included studies to ensure a thorough analysis of the available evidence. After applying a set of inclusion and exclusion criteria, we ultimately included a total of 41 articles in our analysis. To determine the overall effect size for dichotomous variables, we used the Mantel-Haenszel odds ratio in random effect models. For continuous variables, we calculated the inverse variance SMD using random effect models. To assess the outcomes and heterogeneity, we considered p-values (p < 0.05) and I2 values for all outcomes. We performed multivariate and univariate meta-regression analyses using the maximum likelihood approach with the CMA 3.0 software. RESULTS The results of our analysis indicated that the use of ACEIs or ARBs did not significantly influence mortality (OR = 1.10, 95% CI 0.83-1.46, p = 0.43, I2 = 84%), severity (OR = 0.99, 95% CI 0.68-1.45, p = 0.98, I2 = 84%), or healthcare resource utilization (SMD = 0.03, 95% CI 0.06-0.12, p = 0.54, I2 = 37%) in patients with COVID-19 compared to those not taking ACEIs or ARBs. The multivariate meta-regression analysis model explained 63%, 31%, and 100% of the sources of heterogeneity for the three outcome indicators. CONCLUSIONS The use of ACEIs and ARBs is not significantly correlated with mortality, severity, or healthcare resource utilization in patients with COVID-19, indicating safe clinical use of the medications.
Collapse
Affiliation(s)
| | | | - Liang Ren
- Department of Forensic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430032, China; (R.L.)
| |
Collapse
|
8
|
Sheikhi A, Baghaie L, Rahbarizadeh F, Safarzadeh Kozani P, Moradian C, Davidi M, Baharifar N, Kaboli G, Sheikhi M, Li Y, Meghdadi M, Yaish AM, Yu AH, Harless WW, Szewczuk MR. Novel sACE2-Anti-CD16VHH Fusion Protein Surreptitiously Inhibits SARS-CoV-2 Variant Spike Proteins and Macrophage Cytokines, and Activates Natural Killer Cell Cytotoxicity. Vaccines (Basel) 2025; 13:199. [PMID: 40006745 PMCID: PMC11860277 DOI: 10.3390/vaccines13020199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The SARS-CoV-2's high mutations and replication rates contribute to its high infectivity and resistance to current vaccinations and treatments. The primary cause of resistance to most current treatments aligns within the coding regions for the spike S protein of SARS-CoV-2 that has mutated. As a potential novel immunotherapy, we generated a novel fusion protein composed of a soluble ACE2 (sACE2) linked to llama-derived anti-CD16 that targets different variants of spike proteins and enhances natural killer cells to target infected cells. Methods: Here, we generated a novel sACE2-AntiCD16VHH fusion protein using a Gly4Ser linker, synthesized and cloned into the pLVX-EF1alpha-IRES-Puro vector, and further expressed in ExpiCHO-S cells and purified using Ni+NTA chromatography. Results: The fusion protein significantly blocked SARS-CoV-2 alpha, beta, delta, gamma, and omicron S-proteins binding and activating angiotensin-converting enzyme receptor-2 (ACE2) on ACE2-expressing RAW-Blue macrophage cells and the secretion of several key inflammatory cytokines, G-CSF, MIP-1A, and MCP-1, implicated in the cytokine release storm (CRS). The sACE2-Anti-CD16VHH fusion protein also bridged NK cells to ACE2-expressing human lung carcinoma A549 cells and significantly activated NK-dependent cytotoxicity. Conclusions: The findings show that a VHH directed against CD16 could be an excellent candidate to be linked to soluble ACE2 to generate a bi-specific molecule (sACE2-AntiCD16VHH) suitable for bridging effector cells and infected target cells to inhibit SARS-CoV-2 variant spike proteins binding to the ACE2 receptor in the RAW-Blue cell line and pro-inflammatory cytokines and to activate natural killer cell cytotoxicity.
Collapse
Affiliation(s)
- Abdolkarim Sheikhi
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada;
- Department of Immunology, School of Medicine, Dezful University of Medical Sciences, Dezful 64616-43993, Iran; (N.B.); (G.K.)
| | - Leili Baghaie
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-331, Iran; (F.R.); (P.S.K.); (C.M.)
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran 14115-331, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-331, Iran; (F.R.); (P.S.K.); (C.M.)
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran 14115-331, Iran
| | - Cobra Moradian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-331, Iran; (F.R.); (P.S.K.); (C.M.)
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran 14115-331, Iran
| | - Mohammadreza Davidi
- Faculty of Medicine, Kazeroon Azad University, Kazeroon 14778-93855, Iran; (M.D.); (M.S.)
| | - Narges Baharifar
- Department of Immunology, School of Medicine, Dezful University of Medical Sciences, Dezful 64616-43993, Iran; (N.B.); (G.K.)
| | - Golnaz Kaboli
- Department of Immunology, School of Medicine, Dezful University of Medical Sciences, Dezful 64616-43993, Iran; (N.B.); (G.K.)
| | - Mehdi Sheikhi
- Faculty of Medicine, Kazeroon Azad University, Kazeroon 14778-93855, Iran; (M.D.); (M.S.)
| | - Yunfan Li
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada;
| | - Mohammadamin Meghdadi
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (M.M.); (A.M.Y.); (A.H.Y.)
| | - Abdulrahman M. Yaish
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (M.M.); (A.M.Y.); (A.H.Y.)
| | - Aiden H. Yu
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (M.M.); (A.M.Y.); (A.H.Y.)
| | | | - Myron R. Szewczuk
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada;
| |
Collapse
|
9
|
Braga GCD, Ribeiro-Silva JC, Boaro A, Martins FL, Mauad T, Tavares CAM, Teixeira LR, Caramelli B, Girardi ACC. Restoring lung renin-angiotensin system balance through blood pressure control. Clin Sci (Lond) 2025; 139:CS20241155. [PMID: 39905743 DOI: 10.1042/cs20241155] [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: 06/22/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/06/2025]
Abstract
Dysregulated renin-angiotensin system (RAS) signaling contributes to elevated blood pressure (BP), inflammation, and organ damage in systemic arterial hypertension (HTN). We have demonstrated that hypertensive humans and rats exhibit higher expression of classic RAS components and lower expression of counterregulatory RAS components in the lungs compared with normotensive counterparts. Here, we investigated whether BP control could restore the balance between classic [angiotensin I-converting enzyme 2 (ACE)/angiotensin II (Ang II)] and counterregulatory [angiotensin I-converting enzyme 2 (ACE2)/Ang (1-7)] RAS, thereby mitigating lung inflammation. Male spontaneously hypertensive rats (SHRs) were treated with either losartan or amlodipine, both of which effectively reduced BP. These interventions up-regulated lung Ace2 and down-regulated Ace gene expression. Pulmonary membrane ACE2 abundance and activity were higher in losartan- and amlodipine-treated SHRs than in vehicle-treated SHRs, whereas ACE protein and function remained unchanged. Drug-treated SHRs exhibited lower levels of lung Ang II and higher levels of Ang (1-7) than vehicle-treated SHRs. Rebalancing the pulmonary RAS remarkably reduced macrophage number and down-regulated pro-inflammatory genes in SHR lungs, with lower expression of lung pro-inflammatory genes correlating with lower circulating levels of ACE2. Serum analysis in healthy and hypertensive individuals supported these findings, showing higher ACE2 levels in uncontrolled compared with controlled hypertension and normotension. Collectively, these findings suggest that high blood pressure may induce lung inflammation via an ACE/ACE2 imbalance. BP control with either an RAS inhibitor or a calcium channel blocker rebalances RAS in SHR lungs and alleviates inflammation. Furthermore, this study provides a mechanistic link between inflammatory lung diseases (such as COVID-19) and hypertension as a major risk factor.
Collapse
Affiliation(s)
- Gabriela Catão D Braga
- Laboratório de Genética e Cardiologia Molecular, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Joao Carlos Ribeiro-Silva
- Department of Ophthalmology and Visual Sciences, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, USA
| | - Andreia Boaro
- Laboratório de Genética e Cardiologia Molecular, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Flavia Leticia Martins
- Laboratório de Genética e Cardiologia Molecular, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Thais Mauad
- Departmento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Caio A M Tavares
- Unidade de Cardiogeriatria, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Academic Research Organization (ARO), Hospital Israelita Albert Einstein, São Paulo, São Paulo, Brazil
| | - Lisete Ribeiro Teixeira
- Divisão de Pneumologia, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Bruno Caramelli
- Unidade de Medicina Interdisciplinar em Cardiologia, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Adriana C C Girardi
- Laboratório de Genética e Cardiologia Molecular, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
10
|
Xu K, He W, Yu B, Zhong K, Zhou D, Wang DW. Beneficial Effects of Angiotensin II Receptor Blockers on Mortality in Patients with COVID-19: A Retrospective Study from 2019 to 2020 in China. Cardiovasc Drugs Ther 2025; 39:63-74. [PMID: 37566218 DOI: 10.1007/s10557-023-07494-5] [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] [Accepted: 07/23/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND The COVID-19 pandemic has become a serious global public health problem. Although the use of angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin II receptor type 1 blockers (ARBs) has been recommended in patients with COVID-19 and cardiovascular diseases (CVDs), according to the results of some small-sample retrospective analyses, there remains a lack of sufficient evidence to validate their efficacy. This multicenter retrospective study investigated whether ACEI/ARB administration was beneficial in patients with COVID-19 and CVDs. METHODS A total of 11,231 patients with confirmed COVID-19 and CVDs, from 138 hospitals in Hubei Province, were included in this multicenter retrospective study. We compared the clinical characteristics and outcomes between the ARB and non-ARB groups and analyzed the risk factors for in-hospital death using univariate and multivariate Cox regression analyses and Kaplan-Meier curves. RESULTS In the multivariate Cox regression model, after adjusting for age, gender, comorbidities, and in-hospital medications, ARB use was associated with lower all-cause mortality (adjusted HR, 0.53; 95% CI, 0.38-0.73; P < 0.001). After propensity score-matched analysis, the adjusted HR for the use of ARB associated with all-cause mortality was 0.62 (95% CI, 0.40-0.88; P = 0.02). Further subgroup analyses found that the adjusted HRs for the use of ARB associated with all-cause mortality were 0.52 (95% CI, 0.30-0.89; P = 0.016), 0.37 (95% CI, 0.21-0.64; P < 0.001), 0.42 (95% CI, 0.28-0.64; P < 0.001), and 0.55 (95% CI, 0.37-0.84; P = 0.005) in patients with heart failure, diabetes, and hypercholesterolemia, and severe COVID-19, respectively. CONCLUSIONS ARB administration was significantly associated with a lower risk of all-cause mortality in patients with COVID-19 and CVDs. TRIAL REGISTRATION ClinicalTrials.gov NCT05615792. https://www. CLINICALTRIALS gov/ct2/show/NCT05615792.
Collapse
Affiliation(s)
- Ke Xu
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China
| | - Wu He
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China
| | - Bo Yu
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China
| | - Kaineng Zhong
- Health Commission of Hubei Provincial, Wuhan, 430079, China
| | - Da Zhou
- Health Commission of Hubei Provincial, Wuhan, 430079, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China.
| |
Collapse
|
11
|
Lundstrom K. Immunobiology and immunotherapy of COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:73-133. [PMID: 40246352 DOI: 10.1016/bs.pmbts.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The SARS-CoV-2 outbreak in late 2019 triggered a major increase in activities related to immunobiology and immunotherapy to cope with and find solutions to end the COVID-19 pandemic. The unprecedented approach to research and development of drugs and vaccines against SARS-CoV-2 has substantially improved the understanding of immunobiology for COVID-19, which can also be applied to other infectious diseases. Major efforts were dedicated to the repurposing of existing antiviral drugs and the development of novel ones. For this reason, numerous approaches to evaluating interferons, immunoglobulins, and cytokine inhibitors have been conducted. Antibody-based therapies, especially employing monoclonal antibodies have also been on the agenda. Cell-based therapies involving dendritic cells, macrophages, and CAR T-cell approaches have been evaluated. Many existing antiviral drugs have been repurposed for COVID-19 and novel formulations have been tested. The extraordinarily rapid development of efficient vaccines led to the breakthrough of novel vaccine approaches such as mRNA-based vaccines saving millions of lives. Waning immunity of existing vaccines and emerging SARS-CoV-2 variants have required additional booster vaccinations and re-engineering of new versions of COVID-19 vaccines.
Collapse
|
12
|
Zheng X, Xu Z, Xu L, Wang L, Qin S, Ying L, Dong S, Tang L. Angiotensin II Type 2 Receptor Inhibits M1 Polarization and Apoptosis of Alveolar Macrophage and Protects Against Mechanical Ventilation-Induced Lung Injury. Inflammation 2025; 48:165-180. [PMID: 38767784 DOI: 10.1007/s10753-024-02037-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: 02/06/2024] [Revised: 04/03/2024] [Accepted: 04/24/2024] [Indexed: 05/22/2024]
Abstract
Angiotensin II (Ang II) is associated with macrophage polarization and apoptosis, but the role of the angiotensin type 2 receptor (AT2R) in these processes remains controversial. However, the effect of AT2Rs on alveolar macrophages and mechanical ventilation-induced lung injury has not been determined. Mechanical ventilation-induced lung injury in Sprague‒Dawley (SD) rats and LPS-stimulated rat alveolar macrophages (NR8383) were used to determine the effects of AT2Rs, selective AT2R agonists and selective AT1Rs or AT2R antagonists. Macrophage polarization, apoptosis, and related signaling pathways were assessed via western blotting, QPCR and flow cytometry. AT2R expression was decreased in LPS-stimulated rat alveolar macrophages (NR8383). Administration of the AT2R agonist CGP-42112 was associated with an increase in AT2R expression and M2 polarization, but no effect was observed upon administration of the AT2R antagonist PD123319 or the AT1R antagonist valsartan. In mechanical ventilation-induced lung injury in Sprague‒Dawley (SD) rats, the administration of the AT2R agonist C21 was associated with attenuation of the pathological damage score, lung wet/dry weight, cell count and protein content in BALF. C21 can significantly reduce proinflammatory factor TNF-α, IL-1β levels, increase anti-inflammatory factor IL-4, IL-10 levels in BALF, compared with the model group (p < 0.01). Similarly, compared with those at the same time points, the M1/M2 ratios in alveolar macrophages and apoptosis in peritoneal macrophages at 4 h, 6 h and 8 h in the mechanical ventilation models were lower after C21 administration. These findings indicated that the expression of AT2Rs in alveolar macrophages mediates M1 macrophage polarization and apoptosis and that AT2Rs play a protective role in mediating mechanical ventilation-induced lung injury.
Collapse
Affiliation(s)
- Xuyang Zheng
- Department of Pediatrics, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, People's Republic of China.
- Department of Pediatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310000, Zhejiang, People's Republic of China.
| | - Zhiguang Xu
- Department of Pediatrics, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Lihui Xu
- Department of Clinical Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Lingqiao Wang
- Department of Pediatrics, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Siyun Qin
- Department of Pediatrics, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Liu Ying
- Department of Pediatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Shuangyong Dong
- Department of Emergency, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310000, Zhejiang, People's Republic of China.
| | - Lanfang Tang
- Department of pulmonology, Affiliated Children's Hospital, School of medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China.
| |
Collapse
|
13
|
Lin X, Gao F. Angiotensin-Converting Enzyme 2 Enhances Autophagy via the Consumption of miR-326 in a Mouse Model of Acute Lung Injury. Biochem Genet 2025:10.1007/s10528-025-11040-3. [PMID: 39869241 DOI: 10.1007/s10528-025-11040-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/14/2025] [Indexed: 01/28/2025]
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been reported to exert a protective effect in acute lung injury (ALI), though its underlying mechanism remains incompletely understood. In this study, ACE2 expression was found to be upregulated in a mouse model of ALI induced by lipopolysaccharide (LPS) injection. ACE2 knockdown modulated the severity of ALI, the extent of autophagy, and the mTOR pathway in this model. ACE2 regulated liver kinase B1 (LKB1) gene expression by sequestering miR-326, thereby alleviating ALI severity through enhanced autophagy. In cell-based experiments, miR-326 was shown to regulate ACE2 and LKB1 expression and autophagy. Overexpression of ACE2 disrupted miR-326's regulatory effect on LKB1, suggesting that LKB1 may function as an endogenous sponge for miR-326. These findings imply that elevated ACE2 expression in lung could play enhance the autophagy via the consumption of miR-326.
Collapse
Affiliation(s)
- Xingsheng Lin
- Department of Intensive Care Unit, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China
| | - Fengying Gao
- Department of Pulmonary Disease, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| |
Collapse
|
14
|
Zhu Z, Zhang Y, Chen H, Zhang H. Cell-cell crosstalk in the pathogenesis of acute lung injury and acute respiratory distress syndrome. Tissue Barriers 2025:2452082. [PMID: 39798076 DOI: 10.1080/21688370.2025.2452082] [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: 07/08/2024] [Revised: 12/21/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are the result of an exaggerated inflammatory response triggered by a variety of pulmonary and systemic insults. The lung tissues are comprised of a variety of cell types, including alveolar epithelial cells, pulmonary vascular endothelial cells, macrophages, neutrophils, and others. There is mounting evidence that these diverse cell populations within the lung interact to regulate lung inflammation in response to both direct and indirect stimuli. The aim of this review is to provide a summary and discussion of recent advances in the understanding of the importance of cell-cell crosstalk in the pathogenesis of ALI/ARDS, with a specific focus on the cell-cell interactions that may offer prospective therapeutic avenues for ALI/ARDS.
Collapse
Affiliation(s)
- Zhenzhen Zhu
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Ying Zhang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Huan Chen
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Huali Zhang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| |
Collapse
|
15
|
Liu J, Li X, Yang P, He Y, Hong W, Feng Y, Ye Z. LIN28A-dependent lncRNA NEAT1 aggravates sepsis-induced acute respiratory distress syndrome through destabilizing ACE2 mRNA by RNA methylation. J Transl Med 2025; 23:15. [PMID: 39762837 PMCID: PMC11702040 DOI: 10.1186/s12967-024-06032-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a life-threatening and heterogeneous disorder leading to lung injury. To date, effective therapies for ARDS remain limited. Sepsis is a frequent inducer of ARDS. However, the precise mechanisms underlying sepsis-induced ARDS remain unclear. METHODS Here RNA methylation was detected by methylated RNA immunoprecipitation (MeRIP), RNA stability was determined by RNA decay assay while RNA antisense purification (RAP) was used to identify RNA-protein interaction. Besides, co-immunoprecipitation (Co-IP) was utilized to detect protein-protein interaction. Moreover, mice were injected with lipopolysaccharide (LPS) to establish sepsis-induced ARDS model in vivo. RESULTS This study revealed that long non-coding RNA (lncRNA) nuclear-enriched abundant transcript 1 (NEAT1) aggravated lung injury through suppressing angiotensin-converting enzyme 2 (ACE2) in sepsis-induced ARDS models in vitro and in vivo. Mechanistically, NEAT1 declined ACE2 mRNA stability through heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2B1) in lipopolysaccharide (LPS)-treated alveolar type II epithelial cells (AT-II cells). Besides, NEAT1 destabilized ACE2 mRNA depending on RNA methylation by forming methylated NEAT1/hnRNPA2B1/ACE2 mRNA complex in LPS-treated AT-II cells. Moreover, lin-28 homolog A (LIN28A) improved NEAT1 stability whereas insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3) augmented NEAT1 destabilization by associating with LIN28A to disrupt the combination of LIN28A and NEAT1 in LPS-treated AT-II cells. Nevertheless, hnRNPA2B1 increased NEAT1 stability by blocking the interaction between LIN28A and IGF2BP3 in LPS-treated AT-II cells. CONCLUSIONS These findings uncover mechanisms of sepsis-triggering ARDS and provide promising therapeutic targets for sepsis-induced ARDS.
Collapse
Affiliation(s)
- Jun Liu
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Xiang Li
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Peng Yang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yufeng He
- Intensive Care Unit, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Weilong Hong
- Emergency Department, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yawei Feng
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Zhiqiang Ye
- Emergency Department, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
16
|
Bermúdez-Abreut E, Fundora-Barrios T, Hernández Fernández DR, Noa Romero E, Fraga-Quintero A, Casadesús Pazos AV, Fernández-Marrero B, Plasencia Iglesias CA, Clavel Pérez M, Sosa Aguiar K, Sánchez-Ramírez B, Hernández T. Antiviral activity of an ACE2-Fc fusion protein against SARS-CoV-2 and its variants. PLoS One 2025; 20:e0312402. [PMID: 39752453 PMCID: PMC11698409 DOI: 10.1371/journal.pone.0312402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 10/04/2024] [Indexed: 01/06/2025] Open
Abstract
SARS-CoV-2 has continued spreading around the world in recent years since the initial outbreak in 2019, frequently developing into new variants with greater human infectious capacity. SARS-CoV-2 and its mutants use the angiotensin-converting enzyme 2 (ACE2) as a cellular entry receptor, which has triggered several therapeutic strategies against COVID-19 relying on the use of ACE2 recombinant proteins as decoy receptors. In this work, we propose an ACE2 silent Fc fusion protein (ACE2-hFcLALA) as a candidate therapy against COVID-19. This fusion protein was able to block the binding of SARS-CoV-2 RBD to ACE2 receptor as measured by ELISA and flow cytometry inhibition assays. Moreover, we used classical neutralization assays and a progeny neutralization assay to show that the ACE2-hFcLALA fusion protein is capable of neutralizing the authentic virus. Additionally, we found that this fusion protein was more effective in preventing in vitro infection with different variants of interest (alpha, beta, delta, and omicron) compared to the D614G strain. Our results suggest the potential of this molecule to be used in both therapeutic and preventive settings against current and emerging mutants that use ACE2 as a gateway to human cells.
Collapse
Affiliation(s)
| | - Talia Fundora-Barrios
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | | | - Enrique Noa Romero
- National Laboratory of Civil Defense (NLCD), Jamaica Highway and National Highway, San José of Lajas, Mayabeque, Cuba
| | - Anitza Fraga-Quintero
- National Laboratory of Civil Defense (NLCD), Jamaica Highway and National Highway, San José of Lajas, Mayabeque, Cuba
| | - Ana V. Casadesús Pazos
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | | | | | - Marilyn Clavel Pérez
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Katya Sosa Aguiar
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | | | - Tays Hernández
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| |
Collapse
|
17
|
Behzadi P, Chandran D, Chakraborty C, Bhattacharya M, Saikumar G, Dhama K, Chakraborty A, Mukherjee S, Sarshar M. The dual role of toll-like receptors in COVID-19: Balancing protective immunity and immunopathogenesis. Int J Biol Macromol 2025; 284:137836. [PMID: 39613064 DOI: 10.1016/j.ijbiomac.2024.137836] [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: 06/09/2024] [Revised: 11/01/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024]
Abstract
Toll-like receptors (TLRs) of human are considered as the most critical immunological mediators of inflammatory pathogenesis of COVID-19. These immunoregulatory glycoproteins are located on the surface and/or intracellular compartment act as innate immune sensors. Upon binding with distinct SARS-CoV-2 ligand(s), TLRs signal activation of different transcription factors that induce expression of the proinflammatory mediators that collectively induce 'cytokine storm'. Similarly, TLR activation is also pivotal in conferring protection to infection and invasion as well as upregulating the tissue repair pathways. This dual role of the human TLRs in deciding the fate of SARS-CoV-2 has made these receptor proteins as the critical mediators of immunoprotective and immunopathogenic consequences associated with COVID-19. Herein, pathbreaking discoveries exploring the immunobiological importance of the TLRs in COVID-19 and developing TLR-directed therapeutic intervention have been reviewed by accessing the up-to-date literatures available in the public domain/databases. In accordance with our knowledge in association with the importance of TLRs' role against viruses and identification of viral particles, they have been recognized as suitable candidates with high potential as vaccine adjuvants. In this regard, the agonists of TLR4 and TLR9 have effective potential in vaccine technology while the others need further investigations. This comprehensive review suggests that basal level expression of TLRs can act as friends to keep our body safe from strangers but act as a foe via overexpression. Therefore, selective inhibition of the overexpressed TLRs appears to be a solution to counteract the cytokine storm while TLR-agonists as vaccine adjuvants could lessen the risk of infection in the naïve population.
Collapse
Affiliation(s)
- Payam Behzadi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, 37541-374, Iran.
| | | | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, 700126, West Bengal, India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, VyasaVihar, Balasore, 756020, Odisha, India
| | - Guttula Saikumar
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, 243122, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, 243122, India.
| | - Ankita Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, 713340, West Bengal, India
| | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, 713340, West Bengal, India.
| | - Meysam Sarshar
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, 00146, Rome, Italy
| |
Collapse
|
18
|
Ibraheem AAA, Saleh SA, Emam AA, Yousef AA, Abdulhay M, Haridi MK, Wahba AA, Al-Fahham MM, Selim DM, Razek SA, Sorour EI, Abouzied ESHF, Ismail AH, Mohamed SA, Soliman AA, Shehata H, Arab F, Rashad MLM, Hafez SFM, Abdelkhalek K, Ibrahim DM, Ashraf B, Saleh ASE, Fouad RA, Omar WE, Nabil RM, Ramadan RA, El-Sehsah EM, Afify MR, Bawazir Y, Mustafa M, Daghistani Y, Thabit RA, Salah W, Almoraie LM, Aljamei HM, Hummdi LA, Arishi EA, Salem HF, Massoud YM, Khalil DM, Raouf BMA, Elmikaty HA, El-Gaaly SAA, Fakhreldin AR, Hashem MIA. Angiotensin-Converting Enzyme 2 (G8790A) Gene Polymorphism as a Risk Factor for COVID-19 in Egyptian Children and Adolescents. Pediatr Pulmonol 2025; 60:e27479. [PMID: 39821718 DOI: 10.1002/ppul.27479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVE Recently, angiotensin-converting enzyme 2 (ACE2) gene has emerged as a potential candidate gene for susceptibility to SARS-CoV-2 infection. We investigated whether ACE2 G8790A (rs2285666) polymorphism could be a genetic marker for susceptibility to COVID-19 and disease severity in Egyptian children and adolescents. METHODS This was a prospective case-control study included 580 cases diagnosed with COVID-19, and 580 matched control children and adolescents. The ACE2 G8790A (rs2285666) polymorphism was genotyped using polymerase chain reaction (PCR) and ACE2 serum level was measured by ELISA. RESULTS The ACE2 A/A genotype and A-allele were significantly more represented in cases with COVID-19 as compared to control group (44% vs. 30%; OR = 2.83; [95% CI: 1.27-2.63]; p = 0.006; for the A/A genotype) and (65% vs. 51%; OR = 1.9; [95% CI: 1.06-1.72]; p = 0.01; for the A-allele). The presence of ACE2 G/G genotype was an independent risk factor for severe disease (adjusted OR: 2.08; [95% CI: 1.57-6.78]; p = 0.003). CONCLUSION The ACE2 G8790A (rs2285666) polymorphism may confer susceptibility to COVID-19 in Egyptian children and adolescents. The ACE2 G/G genotype and G-allele was associated with lower ACE2 serum levels and may constitute independent risk factors for disease severity.
Collapse
Affiliation(s)
- Ahmed A A Ibraheem
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sarah A Saleh
- Department of Pediatrics, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Ahmed A Emam
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Aly A Yousef
- Department of Pediatrics, Faculty of Medicine, Helwan University, Helwan, Egypt
| | - Mohamed Abdulhay
- Department of Pediatrics, Faculty of Medicine, Helwan University, Helwan, Egypt
| | - Mohammed K Haridi
- Department of Pediatrics, Faculty of Medicine, Assiut University, Asyut, Egypt
| | - Ali A Wahba
- Department of Pediatrics at SSMC, Sheikh Shakhbout Medical City, Abu Dhabi, UAE
| | - Marwa M Al-Fahham
- Department of Pediatrics, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Dalia M Selim
- Department of Pediatrics, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Suzan A Razek
- Department of Pediatrics, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Ehab I Sorour
- Department of Pediatrics, Faculty of Medicine for Boys, Al-Azhar University, Cairo, Egypt
| | - El Sayed H F Abouzied
- Department of Pediatrics, Faculty of Medicine for Boys, Al-Azhar University, Cairo, Egypt
| | - Ahmed H Ismail
- Department of Pediatrics, Faculty of Medicine for Boys, Al-Azhar University, Assiut, Egypt
| | - Soma A Mohamed
- Department of Pediatrics, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Attia A Soliman
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hassan Shehata
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Faika Arab
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Marwa L M Rashad
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sahbaa F M Hafez
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Khalil Abdelkhalek
- Department of Pediatrics, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Dina M Ibrahim
- Department of Pediatrics, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Bassem Ashraf
- Department of Otorhinolaryngology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed S E Saleh
- Department of Otorhinolaryngology, Faculty of Medicine, Benha University, Banha, Egypt
| | - Rania A Fouad
- Department of Medical Biochemistry, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Walaa E Omar
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rehab M Nabil
- Department of Clinical Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Raghdaa A Ramadan
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman M El-Sehsah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Mansoura University, Egypt
| | - Mona R Afify
- Department of Basic Medical Science, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Yasser Bawazir
- Department of Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Mustafa
- Department of Medicine, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Yassir Daghistani
- Department of Medicine, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Rawan A Thabit
- Department of Radiology, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Wed Salah
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Laila M Almoraie
- Department of Family Medicine, University Medical Center, University of Jeddah, Jeddah, Saudi Arabia
| | - Hanan Maas Aljamei
- Department of Biological Science, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Laila Ahmed Hummdi
- Department of Biological Science, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | | | - Hanan F Salem
- Department of Anesthesia, Faculty of Medicine, Benha University, Banha, Egypt
| | - Yasmine M Massoud
- Department of Tropical Medicine, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Dalia M Khalil
- Department of Psychiatry, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Batoul M Abdel Raouf
- Department of Pediatrics, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Hani A Elmikaty
- Department of Pediatrics, National Research Centre, Ad Doqi, Egypt
| | - Sonya A A El-Gaaly
- Department of Internal Medicine, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Ahmed R Fakhreldin
- Department of Pediatrics, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Mustafa I A Hashem
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
19
|
Milad N, Belisario K, MacKillop J, Hirota JA. Dried Cannabis Use, Tobacco Smoking, and COVID-19 Infection: Findings from a Longitudinal Observational Cohort Study. CANNABIS (ALBUQUERQUE, N.M.) 2024; 7:177-192. [PMID: 39781558 PMCID: PMC11705037 DOI: 10.26828/cannabis/2024/000248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Objective The potential impact of cigarette and cannabis smoking on COVID-19 infection outcomes is not well understood. We investigated the association between combustible tobacco use and dried cannabis use with COVID-19 infection in a longitudinal cohort of community adults. Method The sample comprised 1,343 participants, originally enrolled in 2018, who reported their cigarette and cannabis use in 11 assessments over 44 months, until 2022. COVID-19 infection history were self-reported after the onset of the pandemic. Univariate and multivariate logistic regression analyses were performed. The potentially confounding factor of vaccination status was also considered by stratifying data by booster vaccination self-reporting. Results Among 1,343 participants, 820 (61.1%) reported any COVID-19 infection. Dried cannabis use (46.3% of participants, n = 721) was associated with higher self-reporting of 2+ COVID-19 infections (13.3% vs. 7.3% in non-users, p = .0004), while tobacco use (18.5% of participants, n = 248) had no significant effect (13.3% vs. 10.0% in no use group, p = .116). When stratified into single or dual substance use groups, dried cannabis-only use was associated with increased reporting of 1 or 2+ COVID-19 infections compared to substance non-users, while tobacco-only use and dual use groups were not significantly different from non-users. To account for differences in vaccination rates between substance use groups, we found that, among individuals with a COVID-19 booster vaccine, dried cannabis use was still associated with increased reporting of 2+ COVID-19 infections (p = .008). Conclusions Our study suggests that dried cannabis use is associated with a higher likelihood of reporting 2+ COVID-19 infections. Although the study was observational and relied on self-report infection status, our findings support the need for further investigation into the impact of cannabis use on COVID-19 infection, particularly studies employing controlled experimental designs.
Collapse
Affiliation(s)
- Nadia Milad
- Firestone Institute for Respiratory Health - Division of Respirology, Department of Medicine, McMaster University
- McMaster Immunology Research Centre, McMaster University
| | - Kyla Belisario
- Peter Boris Centre for Addictions Research, McMaster University & St. Joseph's Healthcare
- Department of Psychiatry and Behavioural Neurosciences, McMaster University
| | - James MacKillop
- Peter Boris Centre for Addictions Research, McMaster University & St. Joseph's Healthcare
- Department of Psychiatry and Behavioural Neurosciences, McMaster University
| | - Jeremy A Hirota
- Firestone Institute for Respiratory Health - Division of Respirology, Department of Medicine, McMaster University
- McMaster Immunology Research Centre, McMaster University
- Department of Biology, University of Waterloo
- Department of Medicine, University of British Columbia
| |
Collapse
|
20
|
Abbaszadeh H, Kabiri-Rad H, Mohammadi F, Zangoie S, Rajabi-Moghaddam M, Ghafari S, Ziaee M, Javanmard D, Miri-Moghaddam E. The Association Between Genetic Variants in ACE1and ACE2 Genes with Susceptibility to COVID-19 Infection. Biochem Genet 2024; 62:4679-4692. [PMID: 38349438 DOI: 10.1007/s10528-024-10722-8] [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: 09/19/2023] [Accepted: 01/28/2024] [Indexed: 03/27/2024]
Abstract
Angiotensin-converting enzyme 2 (ACE2) receptors facilitate the entry of the causative virus severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) into target cells. Some ACE gene variants have been suggested to be involved in COVID-19 pathogenesis. So, the aim was to assess the association between ACE1 rs4646994 and ACE2 rs2285666 genes polymorphisms and the susceptibility and severity of COVID-19. This case-control study was conducted on 197 patients with COVID-19 and 197 healthy controls. ACE-1 insertion/deletion (I/D) (rs4646994) and ACE2 rs2285666 genes polymorphisms were determined by the amplification refractory mutation system- polymerase chain reaction (ARMS-PCR) technique. The DD genotype of ACE1 I/D polymorphism was associated with increased susceptibility to COVID-19 infection (p = 0.012), whereas the ID genotype of this polymorphism was associated with decreased susceptibility (p = 0.003) (significance level = 0.017). There was no significant association in allele and genotype distribution of ACE2 rs2285666 polymorphism between cases and controls. The ACE1 I/D polymorphism may be considered as a risk factor for COVID-19 susceptibility.
Collapse
Affiliation(s)
- Hamid Abbaszadeh
- Department of Oral and Maxillofacial Pathology, Faculty of Dentistry, Birjand University of Medical Sciences, Birjand, Iran
| | - Hamid Kabiri-Rad
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Fariba Mohammadi
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Soheila Zangoie
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Mahdieh Rajabi-Moghaddam
- Department of Pathology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Shokouh Ghafari
- Cellular and Molecular Research Center, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Masood Ziaee
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Davod Javanmard
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Ebrahim Miri-Moghaddam
- Department of Molecular Medicine, Faculty of Medicine, Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran, 9717853577.
| |
Collapse
|
21
|
Stich M, Magalhães VG, Bürger F, Garbade SF, Jeltsch K, Mohr K, Haddad A, Elling R, Lang P, Rabsteyn A, Jacobsen E, Bode SFN, Müller B, Kräusslich H, Hoffmann GF, Okun JG, Bartenschlager R, Binder M, Janda A, Renk H, Tönshoff B. Elevated Soluble ACE2 Activity in Children and Adults After SARS-CoV-2 Exposure Irrespective of Laboratory-Confirmed Infection. J Med Virol 2024; 96:e70098. [PMID: 39624009 PMCID: PMC11612704 DOI: 10.1002/jmv.70098] [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: 07/22/2024] [Revised: 10/21/2024] [Accepted: 11/15/2024] [Indexed: 12/06/2024]
Abstract
The pivotal role of the cell entry receptor ACE2 for SARS-CoV-2 infection is well-established. When ACE2 is shed from cell surface into plasma as soluble ACE2 (sACE2), it can effectively neutralize SARS-CoV-2. This longitudinal prospective cohort study analyzed sACE2 activity in 1192 participants, aged 4 months to 81 years, 3 and 12 months after SARS-CoV-2 household exposure. Following SARS-CoV-2 exposure, participants exhibited significantly elevated sACE2 activity, irrespective of confirmed infection, with the highest levels observed in exposed children. Longitudinal analysis revealed a decline in sACE2 levels over time, reaching levels comparable to age- and sex-matched pre-pandemic controls. An increase in sACE2 activity was also confirmed in vitro in Calu-3 (human lung) cells within hours of SARS-CoV-2 exposure, providing a direct link between SARS-CoV-2 exposure and elevated sACE2. This study, therefore, challenges the dichotomy of categorizing SARS-CoV-2 exposed participants as infected or not infected solely on currently established diagnostic assays. It demonstrates lasting host responses independent of B- and T-cell memory and may help to keep SARS-CoV-2 infections in balance and contribute to successful virus clearance in children and adults lacking humoral and cellular immune responses following SARS-CoV-2 exposure. Trial Registration: German Registry for Clinical Studies; Identifier: D 00021521.
Collapse
Affiliation(s)
- Maximilian Stich
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
- Heidelberg UniversityMedical Faculty Heidelberg, Department of Infectious Diseases, Molecular VirologyHeidelbergGermany
- German Cancer Research Center (DKFZ)Division Virus‐Associated CarcinogenesisHeidelbergGermany
- German Center for Infection Research (DZIF)Heidelberg Partner SiteHeidelbergGermany
| | - Vladimir Gonçalves Magalhães
- German Cancer Research Center (DKFZ)Division Virus‐Associated Carcinogenesis, Research Group “Dynamics of Early Viral Infection and the Innate Antiviral Response”HeidelbergGermany
| | - Friederike Bürger
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| | - Sven F. Garbade
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| | - Kathrin Jeltsch
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| | - Kerstin Mohr
- German Cancer Research Center (DKFZ)Division Virus‐Associated Carcinogenesis, Research Group “Dynamics of Early Viral Infection and the Innate Antiviral Response”HeidelbergGermany
| | - Anneke Haddad
- Institute for Infection Prevention and ControlUniversity Medical Centre and Faculty of Medicine FreiburgFreiburg im BreisgauGermany
- Center for Pediatrics and Adolescent MedicineUniversity Medical Centre and Faculty of Medicine FreiburgFreiburg im BreisgauGermany
| | - Roland Elling
- Center for Pediatrics and Adolescent MedicineUniversity Medical Centre and Faculty of Medicine FreiburgFreiburg im BreisgauGermany
- Institute for ImmunodeficiencyMedical Center Freiburg, Germany and Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Peter Lang
- University Children's Hospital TübingenDepartment of Hematology/OncologyTübingenGermany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)Partner Site TübingenTübingenGermany
| | - Armin Rabsteyn
- University Children's Hospital TübingenDepartment of Hematology/OncologyTübingenGermany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)Partner Site TübingenTübingenGermany
- Cluster of Excellence iFIT (EXC2180) “Image‐Guided and Functionally Instructed Tumor Therapies”University of TübingenTübingenGermany
| | - Eva‐Maria Jacobsen
- Department of Pediatrics and Adolescent MedicineUlm University Medical CenterUlmGermany
| | - Sebastian F. N. Bode
- Department of Pediatrics and Adolescent MedicineUlm University Medical CenterUlmGermany
| | - Barbara Müller
- Heidelberg UniversityMedical Faculty Heidelberg, Department of Infectious Diseases, VirologyHeidelbergGermany
| | - Hans‐Georg Kräusslich
- German Center for Infection Research (DZIF)Heidelberg Partner SiteHeidelbergGermany
- Heidelberg UniversityMedical Faculty Heidelberg, Department of Infectious Diseases, VirologyHeidelbergGermany
| | - Georg Friedrich Hoffmann
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| | - Jürgen G. Okun
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| | - Ralf Bartenschlager
- Heidelberg UniversityMedical Faculty Heidelberg, Department of Infectious Diseases, Molecular VirologyHeidelbergGermany
- German Cancer Research Center (DKFZ)Division Virus‐Associated CarcinogenesisHeidelbergGermany
- German Center for Infection Research (DZIF)Heidelberg Partner SiteHeidelbergGermany
| | - Marco Binder
- German Cancer Research Center (DKFZ)Division Virus‐Associated Carcinogenesis, Research Group “Dynamics of Early Viral Infection and the Innate Antiviral Response”HeidelbergGermany
| | - Aleš Janda
- Department of Pediatrics and Adolescent MedicineUlm University Medical CenterUlmGermany
| | - Hanna Renk
- University Children's Hospital TübingenDepartment of Pediatric Neurology and Developmental MedicineTübingenGermany
| | - Burkhard Tönshoff
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| |
Collapse
|
22
|
Akerman A, Fichter C, Milogiannakis V, Esneau C, Silva MR, Ison T, Lopez JA, Naing Z, Caguicla J, Amatayakul-Chantler S, Roth N, Manni S, Hauser T, Barnes T, Boss T, Condylios A, Yeang M, Sato K, Bartlett NN, Darley D, Matthews G, Stark DJ, Promsri S, Rawlinson WD, Murrell B, Kelleher AD, Dwyer D, Sintchenko V, Kok J, Ellis S, Marris K, Knight E, Hoad VC, Irving DO, Gosbell I, Brilot F, Wood J, Aggarwal A, Turville SG. Cross-sectional and longitudinal genotype to phenotype surveillance of SARS-CoV-2 variants over the first four years of the COVID-19 pandemic. EBioMedicine 2024; 110:105415. [PMID: 39549677 PMCID: PMC11599457 DOI: 10.1016/j.ebiom.2024.105415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Continued phenotyping and ongoing molecular epidemiology are important in current and future monitoring of emerging SARS-CoV-2 lineages. Herein we developed pragmatic strategies to track the emergence, spread and phenotype of SARS-CoV-2 variants in Australia in an era of decreasing diagnostic PCR testing and focused cohort-based studies. This was aligned to longitudinal studies that span 4 years of the COVID-19 pandemic. METHODS Throughout 2023, we partnered with diagnostic pathology providers and pathogen genomics teams to identify relevant emerging or circulating variants in the New South Wales (NSW) community. We monitored emerging variants through viral culture, growth algorithms, neutralisation responses and changing entry requirements defined by ACE2 and TMPRSS2 receptor use. To frame this in the context of the pandemic stage, we continued to longitudinally track neutralisation responses at the population level using pooled Intravenous Immunoglobulins (IVIG) derived from in excess of 700,000 donations. FINDINGS In antibodies derived from recent individual donations and thousands of donations pooled in IVIGs, we observed continued neutralisation across prior and emerging variants with EG.5.1, HV.1, XCT and JN.1 ranked as the most evasive SARS-CoV-2 variants. Changes in the type I antibody site at Spike positions 452, 455 and 456 were associated with lowered neutralisation responses in XBB lineages. In longitudinal tracking of population immunity spanning three years, we observed continued maturation of neutralisation breadth to all SARS-CoV-2 variants over time. Whilst neutralisation responses initially displayed high levels of imprinting towards Ancestral and early pre-Omicron lineages, this was slowly countered by increased cross reactive breadth to all variants. We predicted JN.1 to have a marked transmission advantage in late 2023 and this eventuated globally at the start of 2024. We could not attribute this advantage to neutralisation resistance but rather propose that this growth advantage arises from the preferential utilisation of ACE2 pools that cannot engage TMPRSS2 at its Collectrin-Like Domain (CLD). INTERPRETATION The emergence of many SARS-CoV-2 lineages documented at the end of 2023 was found to be initially associated with lowered neutralisation responses. This continued to be countered by the gradual maturation of cross-reactive neutralisation responses over time. The later appearance and dominance of the divergent JN.1 lineage cannot be attributed to a lack of neutralisation responses alone, and our data supports that its dominance is a culmination of both lowered neutralisation and changes in ACE2/TMPRSS2 entry preferences. FUNDING This work was primarily supported by Australian Medical Foundation research grants MRF2005760 (ST, GM & WDR), MRF2001684 (ADK and ST) and Medical Research Future Fund Antiviral Development Call grant (WDR), Medical Research Future Fund COVID-19 grant (MRFF2001684, ADK & SGT) and the New South Wales Health COVID-19 Research Grants Round 2 (SGT).
Collapse
Affiliation(s)
- Anouschka Akerman
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Christina Fichter
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Vanessa Milogiannakis
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Camille Esneau
- Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| | - Mariana Ruiz Silva
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Tim Ison
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Joseph A Lopez
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Zin Naing
- Serology and Virology Division (SAViD), NSW HP SEALS, Randwick, Australia
| | - Joanna Caguicla
- Serology and Virology Division (SAViD), NSW HP SEALS, Randwick, Australia
| | | | - Nathan Roth
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Sandro Manni
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Thomas Hauser
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Thomas Barnes
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Tino Boss
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Anna Condylios
- Serology and Virology Division (SAViD), NSW HP SEALS, Randwick, Australia
| | - Malinna Yeang
- Serology and Virology Division (SAViD), NSW HP SEALS, Randwick, Australia
| | - Kenta Sato
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Nathan N Bartlett
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - David Darley
- St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Gail Matthews
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia; St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Damien J Stark
- Molecular Diagnostic Medicine Laboratory, Sydpath, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Susan Promsri
- Histopath, Pinnacle Office Park, Building B, Level 2/4 Drake Ave, Macquarie Park NSW, 2113, Australia
| | | | - Benjamin Murrell
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Anthony D Kelleher
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Dominic Dwyer
- Centre for Infectious Diseases & Microbiology - Public Health and Institute of Clinical Pathology & Medical Research (ICPMR), New South Wales Health Pathology, Westmead, New South Wales, 2145, Australia
| | - Vitali Sintchenko
- Centre for Infectious Diseases & Microbiology - Public Health and Institute of Clinical Pathology & Medical Research (ICPMR), New South Wales Health Pathology, Westmead, New South Wales, 2145, Australia
| | - Jen Kok
- Centre for Infectious Diseases & Microbiology - Public Health and Institute of Clinical Pathology & Medical Research (ICPMR), New South Wales Health Pathology, Westmead, New South Wales, 2145, Australia
| | - Sally Ellis
- New South Wales Ministry of Health, St Leonards, Australia
| | - Kelsi Marris
- New South Wales Ministry of Health, St Leonards, Australia
| | | | - Veronic C Hoad
- Australian Red Cross Lifeblood, Melbourne, Victoria, Australia
| | - David O Irving
- Australian Red Cross Lifeblood, Melbourne, Victoria, Australia
| | - Iain Gosbell
- Australian Red Cross Lifeblood, Melbourne, Victoria, Australia
| | - Fabienne Brilot
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - James Wood
- School of Population Health, UNSW Sydney, Kensington, New South Wales, 2052, Australia
| | - Anupriya Aggarwal
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Stuart G Turville
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
23
|
Legrand M, Khanna AK, Ostermann M, Kotani Y, Ferrer R, Girardis M, Leone M, DePascale G, Pickkers P, Tissieres P, Annoni F, Kotfis K, Landoni G, Zarbock A, Wieruszewski PM, De Backer D, Vincent JL, Bellomo R. The renin-angiotensin-aldosterone-system in sepsis and its clinical modulation with exogenous angiotensin II. Crit Care 2024; 28:389. [PMID: 39593182 PMCID: PMC11590289 DOI: 10.1186/s13054-024-05123-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/05/2024] [Indexed: 11/28/2024] Open
Abstract
Dysregulation of the renin-angiotensin-aldosterone-system (RAAS) in sepsis is a complex and early phenomenon with a likely significant contribution to organ failure and patient outcomes. A better understanding of the pathophysiology and intricacies of the RAAS in septic shock has led to the use of exogenous angiotensin II as a new therapeutic agent. In this review, we report a multinational and multi-disciplinary expert panel discussion on the role and implications of RAAS modulation in sepsis and the use of exogenous angiotensin II. The panel proposed guidance regarding patient selection and treatment options with exogenous angiotensin II which should trigger further research.
Collapse
Affiliation(s)
- Matthieu Legrand
- Department of Anesthesia and Perioperative Care, Division of Critical Care Medicine, University of California San Francisco, 521 Parnassus Avenue, San Francisco, CA, 94143, USA.
| | - Ashish K Khanna
- Department of Anesthesiology, Section on Critical Care Medicine, Wake Forest School of Medicine, Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC, USA.
| | - Marlies Ostermann
- Department of Critical Care, Guy's and St Thomas' Hospital, London, UK
| | - Yuki Kotani
- Department of Intensive Care Medicine, Kameda Medical Center, Kamogawa, Japan
| | - Ricard Ferrer
- Department of Intensive Care, Department of Medicine, SODIR Research Group, VHIR, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Massimo Girardis
- Anesthesia and Intensive Care Department, University Hospital of Modena, University of Modena and Reggio Emilia, Modena, Italy
| | - Marc Leone
- Department of Anesthesiology and Intensive Care Unit, Nord Hospital, Aix Marseille University, Assistance Publique Hôpitaux Universitaires de Marseille, Marseille, France
| | - Gennaro DePascale
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Scienze dell'Emergenza, Anestesiologiche e della Rianimazione, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud UMC Nijmegen, Nijmegen, The Netherlands
| | - Pierre Tissieres
- Pediatric Intensive Care and Neonatal Medicine, Bicêtre Hospital, AP-HP Paris Saclay University, Le Kremlin-Bicêtre, Paris, France
| | - Filippo Annoni
- Department of Intensive Care, Erasme University Hospital, Université Libre de Buxelles, Brussels, Belgium
| | - Katarzyna Kotfis
- Department of Anaesthesiology, Intensive Therapy and Pain Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, Albert-Schweitzer Campus 1, Building A1, 48149, Münster, Germany
| | - Patrick M Wieruszewski
- Department of Pharmacy, Mayo Clinic, Rochester, MN, USA
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
| | - Daniel De Backer
- Department of Intensive Care, CHIREC Hospitals, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme University Hospital, Université Libre de Buxelles, Brussels, Belgium
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital, Melbourne, Australia
- Department of Critical Care, The University of Melbourne, Melbourne, Australia
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia
| |
Collapse
|
24
|
Islam MA, Ford Versypt AN. Mathematical Modeling of Impacts of Patient Differences on Renin-Angiotensin System and Applications to COVID-19 Lung Fibrosis Outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2022.11.06.515367. [PMID: 36380760 PMCID: PMC9665336 DOI: 10.1101/2022.11.06.515367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Patient-specific premorbidity, age, and sex are significant heterogeneous factors that influence the severe manifestation of lung diseases, including COVID-19 fibrosis. The renin-angiotensin system (RAS) plays a prominent role in regulating the effects of these factors. Recent evidence shows patient-specific alterations of RAS homeostasis concentrations with premorbidity and the expression level of angiotensin-converting enzyme 2 (ACE2) during COVID-19. However, conflicting evidence suggests decreases, increases, or no changes in RAS peptides after SARS-CoV-2 infection. In addition, detailed mechanisms connecting the patient-specific conditions before infection to infection-induced RAS alterations are still unknown. Here, a multiscale computational model was developed to quantify the systemic contribution of heterogeneous factors of RAS during COVID-19. Three submodels were connected-an agent-based model for in-host COVID-19 response in the lung tissue, a RAS dynamics model, and a fibrosis dynamics model to investigate the effects of patient-group-specific factors in the systemic alteration of RAS and collagen deposition in the lung. The model results indicated cell death due to inflammatory response as a major contributor to the reduction of ACE and ACE2. In contrast, there were no significant changes in ACE2 dynamics due to viral-bound internalization of ACE2. The model explained possible mechanisms for conflicting evidence of patient-group-specific changes in RAS peptides in previously published studies. Simulated results were consistent with reported RAS peptide values for SARS-CoV-2-negative and SARS-CoV-2-positive patients. RAS peptides decreased for all virtual patient groups with aging in both sexes. In contrast, large variations in the magnitude of reduction were observed between male and female virtual patients in the older and middle-aged groups. The patient-specific variations in homeostasis RAS peptide concentrations of SARS-CoV-2-negative patients also affected the dynamics of RAS during infection. The model results also showed that feedback between RAS signaling and renin dynamics could restore ANGI homeostasis concentration but failed to restore homeostasis values of RAS peptides downstream of ANGI. In addition, the results showed that ACE2 variations with age and sex significantly altered the concentrations of RAS peptides and led to collagen deposition with slight variations depending on age and sex. This model may find further applications in patient-specific calibrations of tissue models for acute and chronic lung diseases to develop personalized treatments.
Collapse
|
25
|
Lima EBDS, Carvalho AFS, Zaidan I, Monteiro AHA, Cardoso C, Lara ES, Carneiro FS, Oliveira LC, Resende F, Santos FRDS, Souza-Costa LP, Chaves IDM, Queiroz-Junior CM, Russo RC, Santos RAS, Tavares LP, Teixeira MM, Costa VV, Sousa LP. Angiotensin-(1-7) decreases inflammation and lung damage caused by betacoronavirus infection in mice. Inflamm Res 2024; 73:2009-2022. [PMID: 39292270 DOI: 10.1007/s00011-024-01948-8] [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: 06/04/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVE Pro-resolving molecules, including the peptide Angiotensin-(1-7) [Ang-(1-7)], have potential adjunctive therapy for infections. Here we evaluate the actions of Ang-(1-7) in betacoronavirus infection in mice. METHODS C57BL/6J mice were infected intranasally with the murine betacoronavirus MHV-3 and K18-hACE2 mice were infected with SARS-CoV-2. Mice were treated with Ang-(1-7) (30 µg/mouse, i.p.) at 24-, 36-, and 48-hours post-infection (hpi) or at 24, 36, 48, 72, and 96 h. For lethality evaluation, one additional dose of Ang-(1-7) was given at 120 hpi. At 3- and 5-days post- infection (dpi) blood cells, inflammatory mediators, viral loads, and lung histopathology were evaluated. RESULTS Ang-(1-7) rescued lymphopenia in MHV-infected mice, and decreased airways leukocyte infiltration and lung damage at 3- and 5-dpi. The levels of pro-inflammatory cytokines and virus titers in lung and plasma were decreased by Ang-(1-7) during MHV infection. Ang-(1-7) improved lung function and increased survival rates in MHV-infected mice. Notably, Ang-(1-7) treatment during SARS-CoV-2 infection restored blood lymphocytes to baseline, decreased weight loss, virus titters and levels of inflammatory cytokines, resulting in improvement of pulmonary damage, clinical scores and lethality rates. CONCLUSION Ang-(1-7) protected mice from lung damage and death during betacoronavirus infections by modulating inflammation, hematological parameters and enhancing viral clearance.
Collapse
Affiliation(s)
- Erick Bryan de Sousa Lima
- Programa de Pós-graduação em Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
- Hospital das Clínicas da Universidade Federal de Minas Gerais/Ebserh, Belo Horizonte, Minas Gerais, Brazil
| | - Antônio Felipe S Carvalho
- Programa de Pós-graduação em Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
- Hospital das Clínicas da Universidade Federal de Minas Gerais/Ebserh, Belo Horizonte, Minas Gerais, Brazil
| | - Isabella Zaidan
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adelson Héric A Monteiro
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila Cardoso
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Edvaldo S Lara
- Programa de Pós-graduação em Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Fernanda S Carneiro
- Programa de Pós-graduação em Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Leonardo C Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Filipe Resende
- Programa de Pós-graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Felipe Rocha da Silva Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiz Pedro Souza-Costa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ian de Meira Chaves
- Programa de Pós-graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz-Junior
- Programa de Pós-graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo C Russo
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Robson A S Santos
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, National Institute in Science and Technology in nanobiopharmaceutics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana P Tavares
- Department of Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian V Costa
- Programa de Pós-graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Programa de Pós-graduação em Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil.
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
26
|
Yu W, Lv Y, Xuan R, Han P, Xu H, Ma X. Human placental mesenchymal stem cells transplantation repairs the alveolar epithelial barrier to alleviate lipopolysaccharides-induced acute lung injury. Biochem Pharmacol 2024; 229:116547. [PMID: 39306309 DOI: 10.1016/j.bcp.2024.116547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/18/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are accompanied by high mortality rates and few effective treatments. Transplantation of human placental mesenchymal stem cells (hPMSCs) may attenuate ALI and the mechanism is still unclear. Our study aimed to elucidate the potential protective effect and therapeutic mechanism of hPMSCs against lipopolysaccharide (LPS)-induced ALI, An ALI model was induced by tracheal instillation of LPS into wild-type (WT) and angiotensin-converting enzyme 2 (ACE2) knockout (KO) male mice, followed by injection of hPMSCs by tail vein. Treatment with hPMSCs improved pulmonary histopathological injury, reduced pulmonary injury scores, decreased leukocyte count and protein levels in bronchoalveolar lavage fluid(BALF), protected the damaged alveolar epithelial barrier, and reversed LPS-induced upregulation of pro-inflammatory factors Interleukin-6 (IL-6) and Tumor necrosis factor-α(TNF-α) and downregulation of anti-inflammatory factor Interleukin-6(IL-10) in BALF. Moreover, administration of hPMSCs inhibited Angiotensin (Ang)II activation and promoted the expression levels of ACE2 and Ang (1-7) in ALI mice. Pathological damage, inflammation levels, and disruption of alveolar epithelial barrier in ALI mice were elevated after the deletion of ACE2 gene, and the Renin angiotensin system (RAS) imbalance was exacerbated. The therapeutic effect of hPMSCs was significantly reduced in ACE2 KO mice. Our findings suggest that ACE2 plays a key role in hPMSCs repairing the alveolar epithelial barrier to protect against ALI, laying a new foundation for the clinical treatment of ALI.
Collapse
Affiliation(s)
- Wenqin Yu
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Ningxia Institute of Human Stem Cells, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China
| | - Yuzhen Lv
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Ningxia Institute of Human Stem Cells, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China
| | - Ruirui Xuan
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China
| | - Peipei Han
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China
| | - Haihuan Xu
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Ningxia Institute of Human Stem Cells, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China
| | - Xiaowei Ma
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China.
| |
Collapse
|
27
|
Houghton MJ, Balland E, Gartner MJ, Thomas BJ, Subbarao K, Williamson G. The flavonoid quercetin decreases ACE2 and TMPRSS2 expression but not SARS-CoV-2 infection in cultured human lung cells. Biofactors 2024; 50:1268-1286. [PMID: 38886986 PMCID: PMC11627474 DOI: 10.1002/biof.2084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/11/2024] [Indexed: 06/20/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) binds to angiotensin-converting enzyme 2 (ACE2) on host cells, via its spike protein, and transmembrane protease, serine 2 (TMPRSS2) cleaves the spike-ACE2 complex to facilitate virus entry. As rate-limiting steps for virus entry, modulation of ACE2 and/or TMPRSS2 may decrease SARS-CoV-2 infectivity and COVID-19 severity. In silico modeling suggested the natural bioactive flavonoid quercetin can bind to ACE2 and a recent randomized clinical trial demonstrated that oral supplementation with quercetin increased COVID-19 recovery. A range of cultured human cells were assessed for co-expression of ACE2 and TMPRSS2. Immortalized Calu-3 lung cells, cultured and matured at an air-liquid interface (Calu-3-ALIs), were established as the most appropriate. Primary bronchial epithelial cells (PBECs) were obtained from healthy adult males (N = 6) and cultured under submerged conditions to corroborate the outcomes. Upon maturation or reaching 80% confluence, respectively, the Calu-3-ALIs and PBECs were treated with quercetin, and mRNA and protein expression were assessed by droplet digital PCR and ELISA, respectively. SARS-CoV-2 infectivity, and the effects of pre- and co-treatment with quercetin, was assessed by median tissue culture infectious dose assay. Quercetin dose-dependently decreased ACE2 and TMPRSS2 mRNA and protein in both Calu-3-ALIs and PBECs after 4 h, while TMPRSS2 remained suppressed in response to prolonged treatment with lower doses (twice daily for 3 days). Quercetin also acutely decreased ADAM17 mRNA, but not ACE, in Calu-3-ALIs, and this warrants further investigation. Calu-3-ALIs, but not PBECs, were successfully infected with SARS-CoV-2; however, quercetin had no antiviral effect, neither directly nor indirectly through downregulation of ACE2 and TMPRSS2. Calu-3-ALIs were reaffirmed to be an optimal cell model for research into the regulation of ACE2 and TMPRSS2, without the need for prior genetic modification, and will prove valuable in future coronavirus and respiratory infectious disease work. However, our data demonstrate that a significant decrease in the expression of ACE2 and TMPRSS2 by a promising prophylactic candidate may not translate to infection prevention.
Collapse
Affiliation(s)
- Michael James Houghton
- Department of Nutrition, Dietetics and FoodMonash University, BASE FacilityNotting HillVICAustralia
- Victorian Heart InstituteMonash University, Victorian Heart HospitalClaytonVICAustralia
| | - Eglantine Balland
- Department of Nutrition, Dietetics and FoodMonash University, BASE FacilityNotting HillVICAustralia
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental BiologyMonash UniversityClaytonVICAustralia
| | - Matthew James Gartner
- Department of Microbiology and ImmunologyUniversity of Melbourne at The Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Belinda Jane Thomas
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
- Monash Lung and Sleep, Monash Health, Monash Medical CentreClaytonVICAustralia
| | - Kanta Subbarao
- Department of Microbiology and ImmunologyUniversity of Melbourne at The Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
- WHO Collaborating Centre for Reference and Research on InfluenzaThe Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Gary Williamson
- Department of Nutrition, Dietetics and FoodMonash University, BASE FacilityNotting HillVICAustralia
- Victorian Heart InstituteMonash University, Victorian Heart HospitalClaytonVICAustralia
| |
Collapse
|
28
|
Sinha S, Callow BW, Farfel AP, Roy S, Chen S, Masotti M, Rajendran S, Buschhaus JM, Espinoza CR, Luker KE, Ghosh P, Luker GD. Breast cancers that disseminate to bone marrow acquire aggressive phenotypes through CX43-related tumor-stroma tunnels. J Clin Invest 2024; 134:e170953. [PMID: 39480488 PMCID: PMC11645149 DOI: 10.1172/jci170953] [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: 03/29/2023] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
Estrogen receptor-positive (ER+) breast cancer commonly disseminates to bone marrow, where interactions with mesenchymal stromal cells (MSCs) shape disease trajectory. We modeled these interactions with tumor-MSC co-cultures and used an integrated transcriptome-proteome-network-analyses workflow to identify a comprehensive catalog of contact-induced changes. Conditioned media from MSCs failed to recapitulate genes and proteins, some borrowed and others tumor-intrinsic, induced in cancer cells by direct contact. Protein-protein interaction networks revealed the rich connectome between "borrowed" and "intrinsic" components. Bioinformatics prioritized one of the borrowed components, CCDC88A/GIV, a multi-modular metastasis-related protein that has recently been implicated in driving a hallmark of cancer, growth signaling autonomy. MSCs transferred GIV protein to ER+ breast cancer cells (that lack GIV) through tunnelling nanotubes via connexin (Cx)43-facilitated intercellular transport. Reinstating GIV alone in GIV-negative breast cancer cells reproduced approximately 20% of both the borrowed and the intrinsic gene induction patterns from contact co-cultures; conferred resistance to anti-estrogen drugs; and enhanced tumor dissemination. Findings provide a multiomic insight into MSC→tumor cell intercellular transport and validate how transport of one such candidate, GIV, from the haves (MSCs) to have-nots (ER+ breast cancer) orchestrates aggressive disease states.
Collapse
Affiliation(s)
- Saptarshi Sinha
- Department of Cellular and Molecular Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | | | | | - Suchismita Roy
- Department of Cellular and Molecular Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | - Siyi Chen
- Center for Molecular Imaging, Department of Radiology
| | | | | | - Johanna M. Buschhaus
- Center for Molecular Imaging, Department of Radiology
- Department of Biomedical Engineering, and
| | - Celia R. Espinoza
- Department of Cellular and Molecular Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | - Kathryn E. Luker
- Center for Molecular Imaging, Department of Radiology
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, School of Medicine, UCSD, La Jolla, California, USA
- Moores Comprehensive Cancer Center
- Department of Medicine
- School of Medicine, and Veterans Affairs Medical Center, UCSD, La Jolla, California, USA
| | - Gary D. Luker
- Center for Molecular Imaging, Department of Radiology
- Department of Biomedical Engineering, and
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
29
|
Boraey NF, Bebars MA, Wahba AA, Abd El Lateef HM, Attia MA, Elsayed AH, Rashed KA, Sorour EI, Ahmed MF, Abd-Elrehim GAB, Soliman AA, Shehab MMM, Elhindawy EM, Ibraheem AAA, Shehata H, Yousif YM, Hashem MIA, Ahmed AA, Emam AA, Gameil DM, Abdelhady EM, Abdelkhalek K, Morsi WEMA, Selim DM, Razek SA, Ashraf B, Saleh ASE, Eltrawy HH, Alanwar MI, Fouad RA, Omar WE, Nabil RM, Abdelhamed MR, Ibrahim MY, Malek MM, Afify MR, Alharbi MT, Nagshabandi MK, Tarabulsi MK, Qashqary ME, Almoraie LM, Salem HF, Rashad MM, El-Gaaly SAA, El-Deeb NA, Abdallah AM, Fakhreldin AR, Hassouba M, Massoud YM, Attaya MSM, Haridi MK. Association of ACE1 I/D polymorphism and susceptibility to COVID-19 in Egyptian children and adolescents. Pediatr Res 2024; 96:1347-1354. [PMID: 38177248 PMCID: PMC11521986 DOI: 10.1038/s41390-023-02982-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/19/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Given the sparse data on the renin-angiotensin system (RAS) and its biological effector molecules ACE1 and ACE2 in pediatric COVID-19 cases, we investigated whether the ACE1 insertion/deletion (I/D) polymorphism could be a genetic marker for susceptibility to COVID-19 in Egyptian children and adolescents. METHODS This was a case-control study included four hundred sixty patients diagnosed with COVID-19, and 460 well-matched healthy control children and adolescents. The I/D polymorphism (rs1799752) in the ACE1 gene was genotyped by polymerase chain reaction (PCR), meanwhile the ACE serum concentrations were assessed by ELISA. RESULTS The ACE1 D/D genotype and Deletion allele were significantly more represented in patients with COVID-19 compared to the control group (55% vs. 28%; OR = 2.4; [95% CI: 1.46-3.95]; for the DD genotype; P = 0.002) and (68% vs. 52.5%; OR: 1.93; [95% CI: 1.49-2.5] for the D allele; P = 0.032). The presence of ACE1 D/D genotype was an independent risk factor for severe COVID-19 among studied patients (adjusted OR: 2.6; [95% CI: 1.6-9.7]; P < 0.001. CONCLUSIONS The ACE1 insertion/deletion polymorphism may confer susceptibility to SARS-CoV-2 infection in Egyptian children and adolescents. IMPACT Recent studies suggested a crucial role of renin-angiotensin system and its biological effector molecules ACE1 and ACE2 in the pathogenesis and progression of COVID-19. To our knowledge, ours is the first study to investigate the association of ACE1 I/D polymorphism and susceptibility to COVID-19 in Caucasian children and adolescents. The presence of the ACE1 D/D genotype or ACE1 Deletion allele may confer susceptibility to SARS-CoV-2 infection and being associated with higher ACE serum levels; may constitute independent risk factors for severe COVID-19. The ACE1 I/D genotyping help design further clinical trials reconsidering RAS-pathway antagonists to achieve more efficient targeted therapies.
Collapse
Affiliation(s)
- Naglaa F Boraey
- Department of Pediatrics, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Marwa A Bebars
- Department of Pediatrics, Princess Alexandra hospital, Harlow, UK
| | - Ali A Wahba
- Department of Pediatrics at SSMC (Sheikh Shakhbout Medical City, Abu Dhabi, UAE
| | | | - Mohamed Atif Attia
- Department of Pediatrics at SKMC (Sheikh khalifa Medical City, Abu Dhabi, UAE
| | - Ahmed H Elsayed
- Department of Pediatrics, Faculty of Medicine for Boys, Al-Azhar University, Al-Azhar, Egypt
| | - Khalid A Rashed
- Department of Pediatrics, Faculty of Medicine for Boys, Al-Azhar University, Al-Azhar, Egypt
| | - Ehab I Sorour
- Department of Pediatrics, Faculty of Medicine for Boys, Al-Azhar University, Al-Azhar, Egypt
| | - Mohamed F Ahmed
- Department of Pediatrics, Faculty of Medicine for Boys, Al-Azhar University, Al-Azhar, Egypt
| | | | - Attia A Soliman
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed M M Shehab
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman M Elhindawy
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed A A Ibraheem
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hassan Shehata
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Yousif M Yousif
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mustafa I A Hashem
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Amani A Ahmed
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed A Emam
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Dalia M Gameil
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman M Abdelhady
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Khalil Abdelkhalek
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Walaa E M A Morsi
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Dalia M Selim
- Department of Pediatrics, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Suzan A Razek
- Department of Pediatrics, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Bassem Ashraf
- Department of Otorhinolaryngology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed S E Saleh
- Department of Otorhinolaryngology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Heba H Eltrawy
- Department of Chest diseases, Faculty of Medicine for Girls, Al-Azhar University, Al-Azhar, Egypt
| | - Mohamed I Alanwar
- Department of Cardiothoracic surgery, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rania A Fouad
- Department of Medical Biochemistry, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Walaa E Omar
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rehab M Nabil
- Department of Clinical Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed R Abdelhamed
- Department of Clinical pathology, Faculty of Medicine for Boys, Al-Azhar University, Al-Azhar, Egypt
| | - Mona Yousri Ibrahim
- Department of Clinical pathology, Faculty of Medicine for Girls, Al-Azhar University, Al-Azhar, Egypt
| | - Mai M Malek
- Department of Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mona R Afify
- Department of Medical microbiology and Parasitology. Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia
| | - Mohanned T Alharbi
- Department of Medical microbiology and Parasitology. Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia
| | - Mohammed K Nagshabandi
- Department of Medical microbiology and Parasitology. Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia
| | - Muyassar K Tarabulsi
- Department of Medical microbiology and Parasitology. Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia
| | - Mohammed Esmail Qashqary
- Department of Family and community medicine, University Medical Center, University of Jeddah, Jeddah, Saudi Arabia
| | - Laila M Almoraie
- Department of Family and community medicine, University Medical Center, University of Jeddah, Jeddah, Saudi Arabia
| | - Hanan F Salem
- Department of Anesthesia, Faculty of Medicine, Benha University, Banha, Egypt
| | - Manal M Rashad
- Department of Anesthesia, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sonya A A El-Gaaly
- Department of Internal Medicine, Faculty of Medicine, Ain-Shams University, Ain-Shams, Egypt
| | - Nahawand A El-Deeb
- Department of Internal Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Amany M Abdallah
- Department of Family Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed R Fakhreldin
- Department of Pediatrics, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Mohamed Hassouba
- Department of Pediatrics, SUNY Downstate Health Science University, Kings County Hospital, Brooklyn, NY, USA
| | - Yasmine M Massoud
- Department of Tropical Medicine, Faculty of Medicine, Ain-Shams University, Ain-Shams, Egypt
| | - Mona S M Attaya
- Department of Pediatrics, Faculty of Medicine for Girls, Al-Azhar University, Al-Azhar, Egypt
| | - Mohammed K Haridi
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
30
|
Ren J, Kitaura H, Noguchi T, Ohori F, Marahleh A, Ma J, Kanou K, Fan Z, Mizoguchi I. Exogenous Angiotensin-(1-7) Provides Protection Against Inflammatory Bone Resorption and Osteoclastogenesis by Inhibition of TNF-α Expression in Macrophages. Calcif Tissue Int 2024; 115:432-444. [PMID: 39030433 PMCID: PMC11405502 DOI: 10.1007/s00223-024-01257-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 07/11/2024] [Indexed: 07/21/2024]
Abstract
Renin-angiotensin-aldosterone system plays a crucial role in the regulation of blood pressure and fluid homeostasis. It is reported to be involved in mediating osteoclastogenesis and bone loss in diseases of inflammatory bone resorption such as osteoporosis. Angiotensin-(1-7), a product of Angiotensin I and II (Ang I, II), is cleaved by Angiotensin-converting enzyme 2 and then binds to Mas receptor to counteract inflammatory effects produced by Ang II. However, the mechanism by which Ang-(1-7) reduces bone resorption remains unclear. Therefore, we aim to elucidate the effects of Ang-(1-7) on lipopolysaccharide (LPS)-induced osteoclastogenesis. In vivo, mice were supracalvarial injected with Ang-(1-7) or LPS ± Ang-(1-7) subcutaneously. Bone resorption and osteoclast formation were compared using micro-computed tomography, tartrate-resistant acid phosphatase (TRAP) stain, and real-time PCR. We found that Ang-(1-7) attenuated tumor necrosis factor (TNF)-α, TRAP, and Cathepsin K expression from calvaria and decreased osteoclast number along with bone resorption at the suture mesenchyme. In vitro, RANKL/TNF-α ± Ang-(1-7) was added to cultures of bone marrow-derived macrophages (BMMs) and osteoclast formation was measured via TRAP staining. The effect of Ang-(1-7) on LPS-induced osteoblasts RANKL expression and peritoneal macrophages TNF-α expression was also investigated. The effect of Ang-(1-7) on the MAPK and NF-κB pathway was studied by Western blotting. As a result, Ang-(1-7) reduced LPS-stimulated macrophages TNF-α expression and inhibited the MAPK and NF-κB pathway activation. However, Ang-(1-7) did not affect osteoclastogenesis induced by RANKL/TNF-α nor reduce osteoblasts RANKL expression in vitro. In conclusion, Ang-(1-7) alleviated LPS-induced osteoclastogenesis and bone resorption in vivo via inhibiting TNF-α expression in macrophages.
Collapse
Affiliation(s)
- Jiayi Ren
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Hideki Kitaura
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan.
| | - Takahiro Noguchi
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Fumitoshi Ohori
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Aseel Marahleh
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | - Jinghan Ma
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Kayoko Kanou
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Ziqiu Fan
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Itaru Mizoguchi
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
31
|
Hashemi ZS, Khalili S, Barough MS, Sarrami Forooshani R, Sanati H, Sarafrazi Esfandabadi F, Rasaee MJ, Nasirmoghadas P. Characterization of an engineered ACE2 protein for its improved biological features and its transduction into MSCs: A novel approach to combat COVID-19 infection. Int J Biol Macromol 2024; 277:134066. [PMID: 39059530 DOI: 10.1016/j.ijbiomac.2024.134066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/06/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
Transduced MSCs that express engineered ACE2 could be highly beneficial to combat COVID-19. Engineered ACE2 can act as decoy targets for the virus, preventing its entry into healthy lung cells. To this end, genetic engineering techniques were used to integrate the ACE2 gene into the MSCs genome. The MSCs were evaluated for proper expression and functionality. The mutated form of ACE2 was characterized using various techniques such as protein expression analysis, binding affinity against spike protein, thermal stability assessment, and enzymatic activity assays. The functionality of the mACE2 was assessed on SARS-CoV-2 using the virus-neutralizing test. The obtained results indicated that by introducing specific mutations in the ACE2 gene, the resulting mutant ACE2 had enhanced interaction with viral spike protein, its thermal stability was increased, and its enzymatic function was inhibited as a decoy receptor. Moreover, the mACE2 protein showed higher efficacy in the neutralization of the SARS-CoV-2. In conclusion, this study proposes a novel approach with potential benefits such as targeted drug delivery and reduced side effects on healthy tissues. These transduced MSCs can also be used in combination with other anti-COVID-19 treatments. Design of similar engineered biomolecules with desired properties could also be used to target other diseases.
Collapse
Affiliation(s)
- Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran.
| | | | | | - Hassan Sanati
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | | | - Mohammad Javad Rasaee
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pourya Nasirmoghadas
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
32
|
Tran KC, Asfar P, Cheng M, Demiselle J, Singer J, Lee T, Sweet D, Boyd J, Walley K, Haljan G, Sharif O, Geri G, Auchabie J, Quenot JP, Lee TC, Tsang J, Meziani F, Lamontagne F, Dubee V, Lasocki S, Ovakim D, Wood G, Turgeon A, Cohen Y, Lebas E, Goudelin M, Forrest D, Teale A, Mira JP, Fowler R, Daneman N, Adhikari NKJ, Gousseff M, Leroy P, Plantefeve G, Rispal P, Courtois R, Winston B, Reynolds S, Birks P, Bienvenu B, Tadie JM, Talarmin JP, Ansart S, Russell JA. Effects of Losartan on Patients Hospitalized for Acute COVID-19: A Randomized Controlled Trial. Clin Infect Dis 2024; 79:615-625. [PMID: 39325643 PMCID: PMC11426262 DOI: 10.1093/cid/ciae306] [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: 02/21/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) down-regulates angiotensin-converting enzyme 2, potentially increasing angiotensin II. We hypothesized that losartan compared to usual care decreases mortality and is safe in patients hospitalized with coronavirus disease 2019 (COVID-19). We aimed to evaluate the effect of losartan versus usual care on 28-day mortality in patients hospitalized for acute COVID-19. METHODS Eligibility criteria included adults admitted for acute COVID-19. Exclusion criteria were hypotension, hyperkalemia, acute kidney injury, and use of angiotensin receptor blockers (ARBs) or angiotensin-converting enzyme inhibitors within 7 days. Participants were randomized to losartan 25-100 mg/day orally for the hospital duration or 3 months or the control arm (usual care) in 29 hospitals in Canada and France. The primary outcome was 28-day mortality. Secondary outcomes were hospital mortality, organ support, and serious adverse events (SAEs). RESULTS The trial was stopped early because of a serious safety concern with losartan. In 341 patients, any SAE and hypotension were significantly higher in the losartan versus usual care groups (any SAE: 39.8% vs 27.2%, respectively, P = .01; hypotension: 30.4% vs 15.3%, respectively, P < .001) in both ward and intensive care patients. The 28-day mortality did not differ between losartan (6.5%) versus usual care (5.9%) (odds ratio, 1.11 [95% confidence interval, .47-2.64]; P = .81), nor did organ dysfunction or secondary outcomes. CONCLUSIONS Caution is needed in deciding which patients to start or continue using ARBs in patients hospitalized with pneumonia to mitigate risk of hypotension, acute kidney injury, and other side effects. ARBs should not be added to care of patients hospitalized for acute COVID-19. CLINICAL TRIALS REGISTRATION NCT04606563.
Collapse
Affiliation(s)
- Karen C Tran
- Division of General Internal Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, Canada
| | - Pierre Asfar
- Service de Médecine Intensive-Réanimation, Centre Hospitalier Universitaire d’Angers, Angers, France
| | - Matthew Cheng
- McGill’s Interdisciplinary Initiative in Infection and Immunity, Divisions of Infectious Diseases and Medical Microbiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Julien Demiselle
- Service de Médecine Intensive-Réanimation, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Joel Singer
- Centre for Health Evaluation and Outcome Science, St Paul's Hospital and University of British Columbia, Vancouver, Canada
| | - Terry Lee
- Centre for Health Evaluation and Outcome Science, St Paul's Hospital and University of British Columbia, Vancouver, Canada
| | - David Sweet
- Division of General Internal Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, Canada
| | - John Boyd
- Division of Critical Care Medicine, and Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, Canada
| | - Keith Walley
- Division of Critical Care Medicine, and Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, Canada
| | - Greg Haljan
- Department of Medicine and Critical Care Medicine, Surrey Memorial Hospital, British Columbia, Canada
| | - Omar Sharif
- Department of Medicine and Critical Care Medicine, Surrey Memorial Hospital, British Columbia, Canada
| | - Guillaume Geri
- Service de Médecine Intensive-Réanimation, Assistance Publique–Hôpitaux de Paris Ambroise Paré, Boulogne, France
| | - Johann Auchabie
- Service de Réanimation Polyvalente, Centre Hospitalier de Cholet
| | - Jean-Pierre Quenot
- Service de Médecine Intensive-Réanimation, Centre Hospitalier Universitaire Dijon, Dijon, France
| | - Todd C Lee
- McGill's Interdisciplinary Initiative in Infection and Immunity, McGill University Health Centre, Montreal, Quebec, Canada
| | - Jennifer Tsang
- Niagara Health, McMaster University, St Catherines, Ontario, Canada
| | - Ferhat Meziani
- Service de Médecine Intensive-Réanimation, Nouvel Hôpital Civil Strasbourg, Strasbourg, France
| | - Francois Lamontagne
- Centre Hospitalier Universitaire de Sherbrooke, University of Sherbrooke, Quebec, Canada
| | - Vincent Dubee
- Service de Maladies Infectieuses, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - Sigismond Lasocki
- Service de Réanimation Chirurgicale, Centre Hospitalier Universitaire Angers, Angers, France
| | - Daniel Ovakim
- Royal Jubilee Hospital, Island Health, Victoria, British Columbia
| | - Gordon Wood
- Royal Jubilee Hospital, Island Health, Victoria, British Columbia
| | - Alexis Turgeon
- Department of Medicine, Centre Hospitalier Universitaire de Québec–Université Laval, Quebec, Canada
| | - Yves Cohen
- Service de Médecine Intensive-Réanimation, Assistance Publique–Hôpitaux de Paris Avicenne, Bobigny, France
| | - Eddy Lebas
- Service de Réanimation Polyvalente, Centre Hospitalier Bretagne-Atlantique, Vannes, France
| | - Marine Goudelin
- Service de Réanimation Polyvalente, Centre Hospitalier Universitaire Limoges, Limoges, France
| | - David Forrest
- Department of Medicine, Nanaimo Regional General Hospital, British Columbia, Canada
| | - Alastair Teale
- Department of Medicine, Nanaimo Regional General Hospital, British Columbia, Canada
| | - Jean-Paul Mira
- Service de Médecine Intensive-Réanimation, Assistance Publique-Hôpitaux de Paris, Cochin, France
| | - Robert Fowler
- Critical Care Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Nick Daneman
- Critical Care Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Neill K J Adhikari
- Critical Care Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Marie Gousseff
- Service de Médecine Interne–Maladies Infectieuses–Hématologie, Centre Hospitalier Bretagne-Atlantique, Vannes, France
| | - Pierre Leroy
- Service de médecine polyvalente et maladies infectieuses, Centre Hospitalier Melun, Melun, France
| | - Gaetan Plantefeve
- Service de Réanimation Polyvalente, Centre Hospitalier Argenteuil, France
| | - Patrick Rispal
- Department of Medicine, Service de médecine interne, Centre Hospitalier Agen, Agen, France
| | - Roxane Courtois
- Service de Médecine post-urgences–Maladies infectieuses, Centre Hospitalier de Cholet, Cholet, France
| | - Brent Winston
- Departments of Critical Care Medicine, Medicine, and Biochemistry and Molecular Biology, Foothills Medical Centre, University of Calgary, Alberta, Canada
| | - Steve Reynolds
- Critical Care Medicine, Royal Columbian Hospital, New Westminster, British Columbia, Canada
- Department of Medicine, Simon Fraser University, Surrey, British Columbia, Canada
| | - Peter Birks
- Critical Care Medicine, Royal Columbian Hospital, New Westminster, British Columbia, Canada
- Department of Medicine, Simon Fraser University, Surrey, British Columbia, Canada
| | - Boris Bienvenu
- Service de médecine interne, Hôpital St Joseph, Marseille, France
| | - Jean-Marc Tadie
- Service de Médecine Intensive-Réanimation et de Maladies Infectieuses, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Jean-Philippe Talarmin
- Service de médecine interne, maladies du sang et infectiologie, Centre Hospitalier de Quimper, Quimper, France
| | - Severine Ansart
- Service de Maladies Infectieuses, Centre Hospitalier Régional Universitaire Brest, Brest, France
| | - James A Russell
- Division of Critical Care Medicine, and Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, Canada
| |
Collapse
|
33
|
Engin MMN, Özdemir Ö. Role of vitamin D in COVID-19 and other viral infections. World J Virol 2024; 13:95349. [PMID: 39323448 PMCID: PMC11401007 DOI: 10.5501/wjv.v13.i3.95349] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/14/2024] [Accepted: 07/19/2024] [Indexed: 08/29/2024] Open
Abstract
Vitamin D is a steroid hormone that is naturally produced in the body or obtained through dietary sources, primarily under the influence of UVB radiation. This essential nutrient has a vital role in numerous physiological processes, encompassing immune function, cell growth, differentiation, insulin regulation, and cardiovascular well-being, along with its pivotal role in sustaining the delicate equilibrium of calcium and phosphate concentrations in the body. Moreover, vitamin D reinforces mucosal defense and bolsters the immune system through immunomodulation, making it a critical component of overall health. Numerous studies have unveiled the profound connection between vitamin D and the predisposition to respiratory tract infections, including well-known viruses such as influenza and the novel severe acute respiratory syndrome coronavirus 2. Vitamin D deficiency has been consistently linked to increased severity of coronavirus disease 2019 (COVID-19) and a heightened risk of mortality among afflicted individuals. Retrospective observational studies have further substantiated these findings, indicating that levels of vitamin D are linked with both the occurrence and severity of COVID-19 cases. Vitamin D has its influence on viral infections through a multitude of mechanisms, such as promoting the release of antimicrobial peptides and fine-tuning the responses of the immune system. Additionally, vitamin D is intertwined with the intricate network of the renin-angiotensin system, suggesting a potential impact on the development of complications related to COVID-19. While further clinical trials and extensive research are warranted, the existing body of evidence strongly hints at the possible use of vitamin D as a valuable tool in the prophylaxis and management of COVID-19 and other viral infectious diseases.
Collapse
Affiliation(s)
| | - Öner Özdemir
- Division of Allergy and Immunology, Department of Pediatrics, Sakarya Research and Training Hospital, Sakarya University, Faculty of Medicine, Sakarya 54100, Türkiye
| |
Collapse
|
34
|
Attiq A, Afzal S, Wahab HA, Ahmad W, Kandeel M, Almofti YA, Alameen AO, Wu YS. Cytokine Storm-Induced Thyroid Dysfunction in COVID-19: Insights into Pathogenesis and Therapeutic Approaches. Drug Des Devel Ther 2024; 18:4215-4240. [PMID: 39319193 PMCID: PMC11421457 DOI: 10.2147/dddt.s475005] [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: 04/22/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Angiotensin-converting enzyme 2 receptors (ACE2R) are requisite to enter the host cells for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). ACE2R is constitutive and functions as a type I transmembrane metallo-carboxypeptidase in the renin-angiotensin system (RAS). On thyroid follicular cells, ACE2R allows SARS-CoV-2 to invade the thyroid gland, impose cytopathic effects and produce endocrine abnormalities, including stiff back, neck pain, muscle ache, lethargy, and enlarged, inflamed thyroid gland in COVID-19 patients. Further damage is perpetuated by the sudden bursts of pro-inflammatory cytokines, which is suggestive of a life-threatening syndrome known as a "cytokine storm". IL-1β, IL-6, IFN-γ, and TNF-α are identified as the key orchestrators of the cytokine storm. These inflammatory mediators upregulate transcriptional turnover of nuclear factor-kappa B (NF-κB), Janus kinase/signal transducer and activator of transcription (JAK/STAT), and mitogen-activated protein kinase (MAPK), paving the pathway for cytokine storm-induced thyroid dysfunctions including euthyroid sick syndrome, autoimmune thyroid diseases, and thyrotoxicosis in COVID-19 patients. Targeted therapies with corticosteroids (dexamethasone), JAK inhibitor (baricitinib), nucleotide analogue (remdesivir) and N-acetyl-cysteine have demonstrated effectiveness in terms of attenuating the severity and frequency of cytokine storm-induced thyroid dysfunctions, morbidity and mortality in severe COVID-19 patients. Here, we review the pathogenesis of cytokine storms and the mechanisms and pathways that establish the connection between thyroid disorder and COVID-19. Moreover, cross-talk interactions of signalling pathways and therapeutic strategies to address COVID-19-associated thyroid diseases are also discussed herein.
Collapse
Affiliation(s)
- Ali Attiq
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Penang, 11800, Malaysia
| | - Sheryar Afzal
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
| | - Habibah A Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Penang, 11800, Malaysia
| | - Waqas Ahmad
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Penang, 11800, Malaysia
| | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrel Sheikh, 6860404, Egypt
| | - Yassir A Almofti
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Department of Biochemistry, Molecular Biology and Bioinformatics, College of Veterinary Medicine, University of Bahri, Khartoum, 12217, Sudan
| | - Ahmed O Alameen
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Department of Physiology, Faculty of Veterinary Medicine, University of Khartoum, Shambat, 13314, Sudan
| | - Yuan Seng Wu
- Sunway Microbiome Centre, School of Medical and Life Sciences, Sunway University, Subang Jaya, Selangor, 47500, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Subang Jaya, Selangor, 47500, Malaysia
| |
Collapse
|
35
|
Mendes RDS, Silva PL, Robba C, Battaglini D, Lopes-Pacheco M, Caruso-Neves C, Rocco PRM. Advancements in understanding the mechanisms of lung-kidney crosstalk. Intensive Care Med Exp 2024; 12:81. [PMID: 39298036 DOI: 10.1186/s40635-024-00672-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024] Open
Abstract
This narrative review delves into the intricate interplay between the lungs and the kidneys, with a focus on elucidating the pathogenesis of diseases influenced by immunological factors, acid-base regulation, and blood gas disturbances, as well as assessing the effects of various therapeutic modalities on these interactions. Key disorders, such as anti-glomerular basement membrane (anti-GBM) disease, the syndrome of inappropriate antidiuretic hormone secretion (SIADH), and Anti-neutrophil Cytoplasmic Antibodies (ANCA) associated vasculitis (AAV), are also examined to shed light on their underlying mechanisms. This review also explores the relationship between acute respiratory distress syndrome (ARDS) and acute kidney injury (AKI), emphasizing how inflammatory mediators can lead to systemic damage and impact multiple organs. In ARDS, fluid overload exacerbates pulmonary edema, while imbalances in blood volume, such as hypovolemia or hypervolemia, can precipitate renal dysfunction. The review highlights how mechanical ventilation strategies can compromise renal blood flow, trigger systemic inflammation, and induce hemodynamic and neurohormonal alterations, all contributing to lung and kidney damage. The impact of extracorporeal membrane oxygenation (ECMO) on lung-kidney interactions is evaluated, highlighting its role in severe respiratory failure and its renal implications. Emerging therapies, such as mesenchymal stem cells and extracellular vesicles, are discussed as promising avenues to mitigate organ damage and enhance outcomes in critically ill patients. Overall, this review offers a nuanced exploration of lung-kidney dynamics, bridging historical insights with contemporary perspectives. It underscores the clinical significance of these interactions in critically ill patients and advocates for integrated management approaches to optimize patient outcomes.
Collapse
Affiliation(s)
- Renata de Souza Mendes
- Department of Nephrology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Nephrology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Chiara Robba
- IRCCS Policlinico San Martino, Genoa, Italy
- Dipertimento di Scienze Chirurgiche Diagnostiche e Integrate, Policlinico San Martino, IRCCS Per l'Oncologia e Neuroscienze, Università degli Studi di Genova, Genoa, Italy
| | - Denise Battaglini
- IRCCS Policlinico San Martino, Genoa, Italy
- Dipertimento di Scienze Chirurgiche Diagnostiche e Integrate, Policlinico San Martino, IRCCS Per l'Oncologia e Neuroscienze, Università degli Studi di Genova, Genoa, Italy
| | - Miquéias Lopes-Pacheco
- Department of Pediatrics, Center for Cystic Fibrosis and Airway Disease Research, Emory University School of Medicine, Atlanta, GA, USA
| | - Celso Caruso-Neves
- Laboratory of Biochemistry and Cellular Biology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
36
|
Nor Rashid N, Amrani L, Alwan A, Mohamed Z, Yusof R, Rothan H. Angiotensin-Converting Enzyme-2 (ACE2) Downregulation During Coronavirus Infection. Mol Biotechnol 2024:10.1007/s12033-024-01277-5. [PMID: 39266903 DOI: 10.1007/s12033-024-01277-5] [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: 10/09/2023] [Accepted: 08/29/2024] [Indexed: 09/14/2024]
Abstract
Angiotensin-converting enzyme-2 (ACE2) downregulation represents a detrimental factor in people with a baseline ACE2 deficiency associated with older age, hypertension, diabetes, and cardiovascular diseases. Human coronaviruses, including HCoV-NL63, SARS-CoV-1, and SARS CoV-2 infect target cells via binding of viral spike (S) glycoprotein to the ACE2, resulting in ACE2 downregulation through yet unidentified mechanisms. This downregulation disrupts the enzymatic activity of ACE2, essential in protecting against organ injury by cleaving and disposing of Angiotensin-II (Ang II), leading to the formation of Ang 1-7, thereby exacerbating the accumulation of Ang II. This accumulation activates the Angiotensin II type 1 receptor (AT1R) receptor, leading to leukocyte recruitment and increased proinflammatory cytokines, contributing to organ injury. The biological impacts and underlying mechanisms of ACE2 downregulation during SARS-CoV-2 infection have not been well defined. Therefore, there is an urgent need to establish a solid theoretical and experimental understanding of the mechanisms of ACE2 downregulation during SARS-CoV-2 entry and replication in the host cells. This review aims to discuss the physiological impact of ACE2 downregulation during coronavirus infection, the relationship between ACE2 decline and virus pathogenicity, and the possible mechanisms of ACE2 degradation, along with the therapeutic approaches.
Collapse
Affiliation(s)
- Nurshamimi Nor Rashid
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Lina Amrani
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | | | - Zulqarnain Mohamed
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Rohana Yusof
- Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia.
| | - Hussin Rothan
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
- Pfizer, Pearl River, NY, USA.
| |
Collapse
|
37
|
Skeeters S, Bagale K, Stepanyuk G, Thieker D, Aguhob A, Chan KK, Dutzar B, Shalygin S, Shajahan A, Yang X, DaRosa PA, Frazier E, Sauer MM, Bogatzki L, Byrnes-Blake KA, Song Y, Azadi P, Tarcha E, Zhang L, Procko E. Modulation of the pharmacokinetics of soluble ACE2 decoy receptors through glycosylation. Mol Ther Methods Clin Dev 2024; 32:101301. [PMID: 39185275 PMCID: PMC11342882 DOI: 10.1016/j.omtm.2024.101301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 07/16/2024] [Indexed: 08/27/2024]
Abstract
The Spike of SARS-CoV-2 recognizes a transmembrane protease, angiotensin-converting enzyme 2 (ACE2), on host cells to initiate infection. Soluble derivatives of ACE2, in which Spike affinity is enhanced and the protein is fused to Fc of an immunoglobulin, are potent decoy receptors that reduce disease in animal models of COVID-19. Mutations were introduced into an ACE2 decoy receptor, including adding custom N-glycosylation sites and a cavity-filling substitution together with Fc modifications, which increased the decoy's catalytic activity and provided small to moderate enhancements of pharmacokinetics following intravenous and subcutaneous administration in humanized FcRn mice. Most prominently, sialylation of native glycans increases exposures by orders of magnitude, and the optimized decoy is therapeutically efficacious in a mouse COVID-19 model. Ultimately, an engineered and highly sialylated decoy receptor produced using methods suitable for manufacture of representative drug substance has high exposure with a 5- to 9-day half-life. Finally, peptide epitopes at mutated sites in the decoys generally have low binding to common HLA class II alleles and the predicted immunogenicity risk is low. Overall, glycosylation is a critical molecular attribute of ACE2 decoy receptors and modifications that combine tighter blocking of Spike with enhanced pharmacokinetics elevate this class of molecules as viable drug candidates.
Collapse
Affiliation(s)
| | - Kamal Bagale
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | - Sergei Shalygin
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Asif Shajahan
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Xu Yang
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | | | | | | | | | | | - Yifan Song
- Cyrus Biotechnology, Seattle, WA 98121, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | | | - Lianghui Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Erik Procko
- Cyrus Biotechnology, Seattle, WA 98121, USA
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
38
|
Martins ALV, Annoni F, da Silva FA, Bolais-Ramos L, de Oliveira GC, Ribeiro RC, Diniz MML, Silva TGF, Pinheiro BD, Rodrigues NA, Dos Santos Matos AH, Motta-Santos D, Campagnole-Santos MJ, Verano-Braga T, Taccone FS, Santos RAS. Angiotensin-(1-7) infusion in COVID-19 patients admitted to the ICU: a seamless phase 1-2 randomized clinical trial. Ann Intensive Care 2024; 14:139. [PMID: 39231898 PMCID: PMC11374945 DOI: 10.1186/s13613-024-01369-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 08/17/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND The coronavirus-related disease (COVID-19) is mainly characterized by a respiratory involvement. The renin-angiotensin system (RAS) has a relevant role in the pathogenesis of COVID-19, as the virus enters host's cells via the angiotensin-converting enzyme 2 (ACE2). METHODS This investigator-initiated, seamless phase 1-2 randomized clinical trial was conceived to test the safety and efficacy of continuous short-term (up to 7 days) intravenous administration of Angiotensin-(1-7) in COVID-19 patients admitted to two intensive care units (ICU). In addition to standard of care, intravenous administration of Angiotensin-(1-7) was started at 5 mcg/Kg day and increased to 10 mcg/Kg day after 24 h (Phase 1; open label trial) or given at 10 mcg/Kg day and continued for a maximum of 7 days or until ICU discharge (Phase 2; double-blind randomized controlled trial). The rate of serious adverse events (SAEs) served as the primary outcome of the study for Phase 1, and the number of oxygen free days (OFDs) by day 28 for Phase 2. RESULTS Between August 2020 and July 2021, when the study was prematurely stopped due to low recruitment rate, 28 patients were included in Phase 1 and 79 patients in Phase 2. Of those, 78 were included in the intention to treat analysis, and the primary outcome was available for 77 patients. During Phase 1, one SAE (i.e., bradycardia) was considered possibly related to the infusion, justifying its discontinuation. In Phase 2, OFDs did not differ between groups (median 19 [0-21] vs. 14 [0-18] days; p = 0.15). When patients from both phases were analyzed in a pooled intention to treat approach (Phase 1-2 trial), OFDs were significantly higher in treated patients, when compared to controls (19 [0-21] vs. 14 [0-18] days; absolute difference -5 days, 95% CI [0-7] p = 0.04). CONCLUSIONS The main findings of our study indicate that continuous intravenous infusion of Angiotensin-(1-7) at 10 mcg/Kg day in COVID-19 patients admitted to the ICU with severe pneumonia is safe. In Phase II intention to treat analysis, there was no significant difference in OFD between groups. Trial Registration ClinicalTrials.gov Identifier: NCT04633772-Registro Brasileiro de Ensaios Clínicos, UTN number: U1111-1255-7167.
Collapse
Affiliation(s)
- Ana Luiza Valle Martins
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Filippo Annoni
- Department of Intensive Care Erasme Hospital, University Hospital of Brussels (HUB), Lennik Road 808, 1070, Brussels, Belgium
| | - Filipe Alex da Silva
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Lucas Bolais-Ramos
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Gisele Capanema de Oliveira
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Renata Cunha Ribeiro
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Mirella Monique Lana Diniz
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Thuanny Granato Fonseca Silva
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Beatriz Dias Pinheiro
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Natália Abdo Rodrigues
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Alana Helen Dos Santos Matos
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Daisy Motta-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Maria José Campagnole-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Thiago Verano-Braga
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Fabio Silvio Taccone
- Department of Intensive Care Erasme Hospital, University Hospital of Brussels (HUB), Lennik Road 808, 1070, Brussels, Belgium
| | - Robson Augusto Souza Santos
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil.
| |
Collapse
|
39
|
Nunes-Souza V, Alenina N, Qadri F, Mosienko V, Santos RAS, Bader M, Rabelo LA. ACE2 Knockout Mice Are Resistant to High-Fat Diet-Induced Obesity in an Age-Dependent Manner. Int J Mol Sci 2024; 25:9515. [PMID: 39273464 PMCID: PMC11394789 DOI: 10.3390/ijms25179515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) presents pleiotropic actions. It hydrolyzes angiotensin I (AngI) and angiotensin II (AngII) into angiotensin-(1-9) (Ang-(1-9)) and angiotensin-(1-7) (Ang-(1-7)), respectively, as well as participates in tryptophan uptake in the gut and in COVID-19 infection. Our aim was to investigate the metabolic effect of ACE2 deletion in young adults and elderly mice under conditions of high calorie intake. Male C57Bl/6 (WT) and ACE2-deficient (ACE2-/y) mice were analyzed at the age of 6 and 12 months under standard diet (StD) and high-fat diet (HFD). Under StD, ACE2-/y showed lower body weight and fat depots, improved glucose tolerance, enhanced insulin sensitivity, higher adiponectin, and lower leptin levels compared to WT. This difference was even more pronounced after HFD in 6-month-old mice, but, interestingly, it was blunted at the age of 12 months. ACE2-/y presented a decrease in adipocyte diameter and lipolysis, which reflected in the upregulation of lipid metabolism in white adipose tissue through the increased expression of genes involved in lipid regulation. Under HFD, both food intake and total energy expenditure were decreased in 6-month-old ACE2-/y mice, accompanied by an increase in liquid intake, compared to WT mice, fed either StD or HFD. Thus, ACE2-/y mice are less susceptible to HFD-induced obesity in an age-dependent manner, as well as represent an excellent animal model of human lipodystrophy and a tool to investigate new treatments.
Collapse
Affiliation(s)
- Valéria Nunes-Souza
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife 50670-901, Brazil
- National Institute of Science and Technology in Nanobiopharmaceutics (Nanobiofar), Belo Horizonte 31270-901, Brazil;
| | - Natalia Alenina
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
| | - Fatimunnisa Qadri
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
| | - Valentina Mosienko
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Robson Augusto Souza Santos
- National Institute of Science and Technology in Nanobiopharmaceutics (Nanobiofar), Belo Horizonte 31270-901, Brazil;
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, 10117 Berlin, Germany
- Institute for Biology, University of Lübeck, 23562 Lübeck, Germany
| | - Luiza Antas Rabelo
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
- National Institute of Science and Technology in Nanobiopharmaceutics (Nanobiofar), Belo Horizonte 31270-901, Brazil;
- Laboratory of Cardiovascular Reactivity, Metabolic Syndrome Center, Institute of Biological Sciences and Health, Federal University of Alagoas, Maceió 57072-900, Brazil
| |
Collapse
|
40
|
Yang G, Khan A, Liang W, Xiong Z, Stegbauer J. Aortic aneurysm: pathophysiology and therapeutic options. MedComm (Beijing) 2024; 5:e703. [PMID: 39247619 PMCID: PMC11380051 DOI: 10.1002/mco2.703] [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: 12/19/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Aortic aneurysm (AA) is an aortic disease with a high mortality rate, and other than surgery no effective preventive or therapeutic treatment have been developed. The renin-angiotensin system (RAS) is an important endocrine system that regulates vascular health. The ACE2/Ang-(1-7)/MasR axis can antagonize the adverse effects of the activation of the ACE/Ang II/AT1R axis on vascular dysfunction, atherosclerosis, and the development of aneurysms, thus providing an important therapeutic target for the prevention and treatment of AA. However, products targeting the Ang-(1-7)/MasR pathway still lack clinical validation. This review will outline the epidemiology of AA, including thoracic, abdominal, and thoracoabdominal AA, as well as current diagnostic and treatment strategies. Due to the highest incidence and most extensive research on abdominal AA (AAA), we will focus on AAA to explain the role of the RAS in its development, the protective function of Ang-(1-7)/MasR, and the mechanisms involved. We will also describe the roles of agonists and antagonists, suggest improvements in engineering and drug delivery, and provide evidence for Ang-(1-7)/MasR's clinical potential, discussing risks and solutions for clinical use. This study will enhance our understanding of AA and offer new possibilities and promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Guang Yang
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Institute of Translational Medicine Shenzhen Second People's Hospital The First Affiliated Hospital of Shenzhen University Shenzhen China
- Department of Life Sciences Yuncheng University Yuncheng China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Abbas Khan
- Department of Nutrition and Health Promotion University of Home Economics Lahore Pakistan Lahore Pakistan
| | - Wei Liang
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Zibo Xiong
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Johannes Stegbauer
- Department of Nephrology Medical Faculty University Hospital Düsseldorf Heinrich Heine University Düsseldorf Düsseldorf Germany
| |
Collapse
|
41
|
Iwasaki M, Yamamoto M, Tomihari M, Ishikawa M. Ropivacaine Administration Suppressed A549 Lung Adenocarcinoma Cell Proliferation and Migration via ACE2 Upregulation and Inhibition of the Wnt1 Pathway. Int J Mol Sci 2024; 25:9334. [PMID: 39273283 PMCID: PMC11395614 DOI: 10.3390/ijms25179334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Previous studies have suggested that perioperative anesthesia could have direct impacts on cancer cell biology. The present study investigated the effects of ropivacaine administration on lung adenocarcinoma cells. METHODS Ropivacaine was administered to A549 cells at concentrations of 0.1, 1, and 6 mM for 2 h. Angiotensin-converting enzyme 2 (ACE2) small interfering RNA (siRNA) transfection was performed 6 h prior to ropivacaine administration. Cell proliferation and migration were assessed with cell counting kit 8 (CCK-8) and a wound healing assay at 0 and 24 h after anesthesia exposure. PCR arrays were performed, followed by PCR validation. RESULTS Ropivacaine administration inhibited A549 cell proliferation and migration in a concentration-dependent manner, with ACE2 upregulation and HIF1α (hypoxia-inducible factor 1α) downregulation. The anticancer effect of ropivacaine was canceled out via ACE2 siRNA transfection. PCR arrays showed specific gene change patterns in the ropivacaine and respective ACE2-knockdown groups. EGFR (epidermal growth factor receptor), BAX (Bcl-2-associated X protein) and BCL2 (B-cell/CLL lymphoma 2) were suppressed with ropivacaine administration; these effects were reversed via ACE2 siRNA induction. CONCLUSION Ropivacaine administration inhibited A549 cell biology in conjunction with ACE2 upregulation via the inhibition of the Wnt1 (wingless/Integrated 1) pathway.
Collapse
Affiliation(s)
- Masae Iwasaki
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan; (M.Y.); (M.T.); (M.I.)
| | | | | | | |
Collapse
|
42
|
Sinha S, Callow BW, Farfel AP, Roy S, Chen S, Rajendran S, Buschhaus JM, Espinoza CR, Luker KE, Ghosh P, Luker GD. Breast Cancers That Disseminate to Bone Marrow Acquire Aggressive Phenotypes through CX43-related Tumor-Stroma Tunnels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.18.533175. [PMID: 36993616 PMCID: PMC10055300 DOI: 10.1101/2023.03.18.533175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Estrogen receptor-positive (ER+) breast cancer commonly disseminates to bone marrow, where interactions with mesenchymal stromal cells (MSCs) shape disease trajectory. We modeled these interactions with tumor-MSC co-cultures and used an integrated transcriptome-proteome-network-analyses workflow to identify a comprehensive catalog of contact-induced changes. Conditioned media from MSCs failed to recapitulate genes and proteins, some borrowed and others tumor-intrinsic, induced in cancer cells by direct contact. Protein-protein interaction networks revealed the rich connectome between 'borrowed' and 'intrinsic' components. Bioinformatics prioritized one of the 'borrowed' components, CCDC88A /GIV, a multi-modular metastasis-related protein that has recently been implicated in driving a hallmark of cancer, growth signaling autonomy. MSCs transferred GIV protein to ER+ breast cancer cells (that lack GIV) through tunnelling nanotubes via connexin (Cx)43-facilitated intercellular transport. Reinstating GIV alone in GIV-negative breast cancer cells reproduced ∼20% of both the 'borrowed' and the 'intrinsic' gene induction patterns from contact co-cultures; conferred resistance to anti-estrogen drugs; and enhanced tumor dissemination. Findings provide a multiomic insight into MSC→tumor cell intercellular transport and validate how transport of one such candidate, GIV, from the haves (MSCs) to have-nots (ER+ breast cancer) orchestrates aggressive disease states.
Collapse
|
43
|
Zhang Y, Wang D, Wang X, Ma H, Liu Y, Hong Z, Zhu Z, Chen X, Lv D, Cao Y, Chai Y. A dual-target SPR screening system for simultaneous ligand discovery of SARS-CoV-2 spike protein and its receptor ACE2 from Chinese herbs. J Pharm Biomed Anal 2024; 245:116142. [PMID: 38631070 DOI: 10.1016/j.jpba.2024.116142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/14/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024]
Abstract
Traditional Chinese Medicine (TCM) is a supremely valuable resource for the development of drug discovery. Few methods are capable of hunting for potential molecule ligands from TCM towards more than one single protein target. In this study, a novel dual-target surface plasmon resonance (SPR) biosensor was developed to perform targeted compound screening of two key proteins involved in the cellular invasion process of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2): the spike (S) protein receptor binding domain (RBD) and the angiotensin-converting enzyme 2 (ACE2). The screening and identification of active compounds from six Chinese herbs were conducted taking into consideration the multi-component and multi-target nature of Traditional Chinese Medicine (TCM). Puerarin from Radix Puerariae Lobatae was discovered to exhibit specific binding affinity to both S protein RBD and ACE2. The results highlight the efficiency of the dual-target SPR system in drug screening and provide a novel approach for exploring the targeted mechanisms of active components from Chinese herbs for disease treatment.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Biochemical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Dongyao Wang
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Department of Pharmaceutical Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Xiying Wang
- Suzhou Innovation Center of Shanghai University, Suzhou 215127, China
| | - Huilin Ma
- Department of Biochemical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Yue Liu
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Department of Pharmaceutical Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Zhanying Hong
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Department of Pharmaceutical Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Zhenyu Zhu
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Center for Instrumental Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Xiaofei Chen
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Center for Instrumental Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Diya Lv
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Center for Instrumental Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| | - Yan Cao
- Department of Biochemical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| | - Yifeng Chai
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Department of Pharmaceutical Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
44
|
Wu D, Liao X, Gao J, Gao Y, Li Q, Gao W. Potential pharmaceuticals targeting neuroimmune interactions in treating acute lung injury. Clin Transl Med 2024; 14:e1808. [PMID: 39129233 PMCID: PMC11317502 DOI: 10.1002/ctm2.1808] [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: 04/26/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND AND MAIN BODY Although interactions between the nervous and immune systems have been recognized decades ago, it has become increasingly appreciated that neuroimmune crosstalk is among the driving factors of multiple pulmonary inflammatory diseases including acute lung injury (ALI). Here, we review the current understanding of nerve innervations towards the lung and summarize how the neural regulation of immunity and inflammation participates in the onset and progression of several lung diseases, especially ALI. We then present advancements in the development of potential drugs for ALI targeting neuroimmune interactions, including cholinergic anti-inflammatory pathway, sympathetic-immune pathway, purinergic signalling, neuropeptides and renin-angiotensin system at different stages from preclinical investigation to clinical trials, including the traditional Chinese medicine. CONCLUSION This review highlights the importance of considering the therapeutic strategy of inflammatory diseases within a conceptual framework that integrates classical inflammatory cascade and neuroimmune circuits, so as to deepen the understanding of immune modulation and develop more sophisticated approaches to treat lung diseases represented by ALI. KEY POINTS The lungs present abundant nerve innervations. Neuroimmune interactions exert a modulatory effect in the onset and progression of lung inflammatory diseases, especially acute lung injury. The advancements of potential drugs for ALI targeting neuroimmune crosstalk at different stages from preclinical investigation to clinical trials are elaborated. Point out the direction for the development of neuroimmune pharmacology in the future.
Collapse
Affiliation(s)
- Di Wu
- Department of Pulmonary and Critical Care MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Ximing Liao
- Department of Pulmonary and Critical Care MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Jing Gao
- Department of Pulmonary and Critical Care MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Yixuan Gao
- Department of GynaecologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanP. R. China
| | - Qiang Li
- Department of Pulmonary and Critical Care MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Wei Gao
- Department of Pulmonary and Critical Care MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghaiP. R. China
| |
Collapse
|
45
|
Akcilar R, Kocak FE, Kar F, Isiklar OO, Atlanoglu S, Genc O, Yaman F. Evaluation of the relationship between ACE2 G8790A and AT2R A1675G gene polymorphisms in COVID-19 patients with and without lung involvement. ASIAN BIOMED 2024; 18:157-170. [PMID: 39309472 PMCID: PMC11414776 DOI: 10.2478/abm-2024-0022] [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] [Indexed: 09/25/2024]
Abstract
Background The SARS-CoV-2 virus produces severe acute respiratory syndrome. The severity of coronavirus disease 2019 (COVID-19) infection is determined by a number of factors, including inherited ones. Objectives Our goal is to investigate the link between ACE2 G8790A (rs2285666) and AT2R A1675G (rs14035430) gene polymorphisms in COVID-19 patients with and without lung involvement. Methods A total of 160 COVID-19 patients were divided into 2 groups based on their clinical symptoms: those without lung involvement (control group) and those with lung involvement (infected group). The ACE2 G8790A and AT2R A1675G gene polymorphisms were analyzed using the PCR-RFLP methods. Results The GG genotype, G allele of ACE2 G8790A, and GG genotype of AT2R A1675G were significantly higher in the control group and had a protective effect against COVID-19 as well as decreased the development of lung involvement (OR = 0.29, 95% CI = 0.10-0.84; OR = 0.40, 95% CI = 0.22-0.72; and OR = 0.33, 95% CI = 0.14-0.78, respectively). Moreover, we found that the AA genotype, A allele of ACE2 G8790A, and AG genotype of AT2R A1675G increased the risk of COVID-19 in the infected group (OR = 3.50, 95% CI = 1.18-10.3; OR = 2.49, 95% CI = 1.39-4.48; and OR = 3.08, 95% CI = 1.28-7.38, respectively). Conclusions These results revealed that a greater frequency of COVID-19 lung involvement in the Turkish population was connected with the AA genotype, the A allele of ACE2 G8790A, and the AG genotype of AT2R A1675G.
Collapse
Affiliation(s)
- Raziye Akcilar
- Department of Physiology, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| | - Fatma Emel Kocak
- Department of Medical Biochemistry, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| | - Fatih Kar
- Department of Basic Sciences, Kütahya Health Sciences University, Faculty of Natural and Engineering Sciences, Kutahya43100, Turkey
| | - Ozben Ozden Isiklar
- Department of Medical Biochemistry, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| | - Sahinde Atlanoglu
- Department of Radiology, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| | - Ozlem Genc
- Department of Medical Microbiology, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| | - Fatima Yaman
- Department of Physical Medicine and Rehabilitation, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| |
Collapse
|
46
|
Daneshwar D, Lee Y, Nordin A. COVID-19 and Prostatitis: A Review of Current Evidence. Diseases 2024; 12:157. [PMID: 39057128 PMCID: PMC11276594 DOI: 10.3390/diseases12070157] [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: 04/23/2024] [Revised: 05/20/2024] [Accepted: 06/16/2024] [Indexed: 07/28/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19), a highly contagious viral disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), poses a global health threat. The virus enters host cells by binding with angiotensin-converting enzyme 2 (ACE2), which is then facilitated by the protease activity of transmembrane serine protease 2 (TMPRSS2). It triggers a cytokine storm that eventually leads to cell apoptosis, tissue damage, and organ failure. Therefore, any organs in the human body that have both receptors are highly susceptible to COVID-19 infection, potentially resulting in multiple-organ failure. The prostate has been reported to express high levels of ACE2 and TMPRSS2. While there are limited studies regarding the association between COVID-19 and prostatitis, the possibility that SARS-CoV-2 could cause prostatitis cannot be denied. Thus, through this review, a better insight into the associations of SAR-CoV-2 can be provided.
Collapse
Affiliation(s)
- Datesh Daneshwar
- Urology Clinic, Prince Court Medical Centre, 39, Jalan Kia Peng, Kuala Lumpur 50450, Malaysia
| | - Yemin Lee
- MedCentral Consulting, International Youth Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Abid Nordin
- Graduate School of Medicine, KPJ Healthcare University, Nilai 71800, Negeri Sembilan, Malaysia;
| |
Collapse
|
47
|
Grune J, Bajpai G, Ocak PT, Kaufmann E, Mentkowski K, Pabel S, Kumowski N, Pulous FE, Tran KA, Rohde D, Zhang S, Iwamoto Y, Wojtkiewicz GR, Vinegoni C, Green U, Swirski FK, Stone JR, Lennerz JK, Divangahi M, Hulsmans M, Nahrendorf M. Virus-Induced Acute Respiratory Distress Syndrome Causes Cardiomyopathy Through Eliciting Inflammatory Responses in the Heart. Circulation 2024; 150:49-61. [PMID: 38506045 PMCID: PMC11216864 DOI: 10.1161/circulationaha.123.066433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/15/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Viral infections can cause acute respiratory distress syndrome (ARDS), systemic inflammation, and secondary cardiovascular complications. Lung macrophage subsets change during ARDS, but the role of heart macrophages in cardiac injury during viral ARDS remains unknown. Here we investigate how immune signals typical for viral ARDS affect cardiac macrophage subsets, cardiovascular health, and systemic inflammation. METHODS We assessed cardiac macrophage subsets using immunofluorescence histology of autopsy specimens from 21 patients with COVID-19 with SARS-CoV-2-associated ARDS and 33 patients who died from other causes. In mice, we compared cardiac immune cell dynamics after SARS-CoV-2 infection with ARDS induced by intratracheal instillation of Toll-like receptor ligands and an ACE2 (angiotensin-converting enzyme 2) inhibitor. RESULTS In humans, SARS-CoV-2 increased total cardiac macrophage counts and led to a higher proportion of CCR2+ (C-C chemokine receptor type 2 positive) macrophages. In mice, SARS-CoV-2 and virus-free lung injury triggered profound remodeling of cardiac resident macrophages, recapitulating the clinical expansion of CCR2+ macrophages. Treating mice exposed to virus-like ARDS with a tumor necrosis factor α-neutralizing antibody reduced cardiac monocytes and inflammatory MHCIIlo CCR2+ macrophages while also preserving cardiac function. Virus-like ARDS elevated mortality in mice with pre-existing heart failure. CONCLUSIONS Our data suggest that viral ARDS promotes cardiac inflammation by expanding the CCR2+ macrophage subset, and the associated cardiac phenotypes in mice can be elicited by activating the host immune system even without viral presence in the heart.
Collapse
Affiliation(s)
- Jana Grune
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité, Berlin, Germany (J.G.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Physiology, Germany (J.G.)
- German Center for Cardiovascular Research, Partner Site Berlin (J.G.)
| | - Geetika Bajpai
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Pervin Tülin Ocak
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Cardiology, University Hospital Heidelberg, Germany (P.T.O.)
| | - Eva Kaufmann
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, Research Institute McGill University Health Centre, and McGill International TB Centre Montreal, Canada (E.K., K.A.T., M.D.)
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada (E.K.)
| | - Kyle Mentkowski
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Steffen Pabel
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Internal Medicine II, University Medical Center Regensburg, Germany (S.P.)
| | - Nina Kumowski
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Internal Medicine I, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule Aachen University, Germany (N.K.)
| | - Fadi E Pulous
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Kim A Tran
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, Research Institute McGill University Health Centre, and McGill International TB Centre Montreal, Canada (E.K., K.A.T., M.D.)
| | - David Rohde
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Shuang Zhang
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Yoshiko Iwamoto
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Gregory R Wojtkiewicz
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Claudio Vinegoni
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Ursula Green
- Department of Pathology, Center for Integrated Diagnostics (U.G., J.K.L.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Filip K Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY (F.K.S.)
| | - James R Stone
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA (J.R.S.)
- Massachusetts General Hospital, Boston (J.R.S.)
| | - Jochen K Lennerz
- Department of Pathology, Center for Integrated Diagnostics (U.G., J.K.L.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Maziar Divangahi
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, Research Institute McGill University Health Centre, and McGill International TB Centre Montreal, Canada (E.K., K.A.T., M.D.)
| | - Maarten Hulsmans
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Matthias Nahrendorf
- Center for Systems Biology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., Y.I., G.R.W., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Radiology (J.G., G.B., P.T.O., K.M., S.P., N.K., F.E.P., D.R., S.Z., C.V., M.H., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
- Gordon Center for Medical Imaging (M.N.)
- Department of Internal Medicine, University Hospital Wuerzburg, Germany (M.N.)
| |
Collapse
|
48
|
Erinoso O, Osibogun O, Balakrishnan S, Yang W. Long COVID among US adults from a population-based study: Association with vaccination, cigarette smoking, and the modifying effect of chronic obstructive pulmonary disease (COPD). Prev Med 2024; 184:108004. [PMID: 38754738 PMCID: PMC11148848 DOI: 10.1016/j.ypmed.2024.108004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVE Post-COVID Conditions (or Long COVID) have been widely reported, but population-based studies exploring the relationship between its risk factors are lacking. We examined the associations between Long COVID, chronic obstructive pulmonary disease [COPD], vaccination status, and cigarette smoking. We also tested for the modifying effect of COPD status. METHODS Data from the 2022 US nationwide Behavioral Risk Factor Surveillance System (BRFSS) were analyzed. Our primary outcome was Long COVID (Yes/No) after a positive COVID-19 diagnosis. Predictor variables were COPD, coronary heart disease (CHD), diabetes, asthma, body mass index, cigarette smoking status, and number of COVID-19 vaccinations (0-4). Weighted multivariable logistic regression models were used and adjusted for sociodemographic factors. Regression models were used to explore the modifying effects of COPD status. RESULTS The weighted prevalence of Long COVID among survivors (N = 121,379) was 21.8% (95%CI: 21.4, 22.3), with tiredness/fatigue (26.2% [95%:25.1, 27.2]) as the most common symptom. Respondents with COPD (aOR: 1.71 [95%CI: 1.45, 2.02]), current daily smokers (aOR: 1.23 [95%CI:1.01, 1.49]), and former smokers (aOR: 1.24 [95%CI:1.12, 1.38]) (vs. never established smokers) had higher odds of Long COVID. However, respondents who had received three (aOR: 0.75 [95%CI:0.65, 0.85]) and four (aOR: 0.71 [95%CI:0.58, 0.86]) vaccine doses (vs. no vaccine) had lower odds of Long COVID. COPD had a modifying effect on the relationship between cigarette smoking and Long COVID (p-value: 0.013). CONCLUSION Our findings underscore a complex interaction between COPD, cigarette smoking, and Long COVID. Further, COVID-19 vaccination may be protective against Long COVID.
Collapse
Affiliation(s)
- Olufemi Erinoso
- Department of Health Behavior, Policy, and Administration Science, School of Public Health, University of Nevada, Reno, NV, USA.
| | - Olatokunbo Osibogun
- Department of Epidemiology, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA
| | - Siva Balakrishnan
- Department of Epidemiology and Biostatistics, School of Public Health, University of Nevada, Reno, NV, USA
| | - Wei Yang
- Department of Epidemiology and Biostatistics, School of Public Health, University of Nevada, Reno, NV, USA
| |
Collapse
|
49
|
Ameratunga R, Jordan A, Lehnert K, Leung E, Mears ER, Snell R, Steele R, Woon ST. SARS-CoV-2 evolution has increased resistance to monoclonal antibodies and first-generation COVID-19 vaccines: Is there a future therapeutic role for soluble ACE2 receptors for COVID-19? Antiviral Res 2024; 227:105894. [PMID: 38677595 DOI: 10.1016/j.antiviral.2024.105894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
COVID-19 has caused calamitous health, economic and societal consequences. Although several COVID-19 vaccines have received full authorization for use, global deployment has faced political, financial and logistical challenges. The efficacy of first-generation COVID-19 vaccines is waning and breakthrough infections are allowing ongoing transmission and evolution of SARS-CoV-2. Furthermore, COVID-19 vaccine efficacy relies on a functional immune system. Despite receiving three primary doses and three or more heterologous boosters, some immunocompromised patients may not be adequately protected by COVID-19 vaccines and remain vulnerable to severe disease. The evolution of new SARS-CoV-2 variants has also resulted in the rapid obsolescence of monoclonal antibodies. Convalescent plasma from COVID-19 survivors has produced inconsistent results. New drugs such as Paxlovid (nirmatrelvir/ritonavir) are beyond the reach of low- and middle-income countries. With widespread use of Paxlovid, it is likely nirmatrelvir-resistant clades of SARS-CoV-2 will emerge in the future. There is thus an urgent need for new effective anti-SARS-CoV-2 treatments. The in vitro efficacy of soluble ACE2 against multiple SARS-CoV-2 variants including omicron (B.1.1.529), was recently described using a competitive ELISA assay as a surrogate marker for virus neutralization. This indicates soluble wild-type ACE2 receptors are likely to be resistant to viral evolution. Nasal and inhaled treatment with soluble ACE2 receptors has abrogated severe disease in animal models of COVID-19. There is an urgent need for clinical trials of this new class of antiviral therapeutics, which could complement vaccines and Paxlovid.
Collapse
Affiliation(s)
- Rohan Ameratunga
- Department of Clinical Immunology, Auckland Hospital, Park Rd, Grafton, 1010, Auckland, New Zealand; Department of Virology and Immunology, Auckland Hospital, Park Rd, Grafton, 1010, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Anthony Jordan
- Department of Clinical Immunology, Auckland Hospital, Park Rd, Grafton, 1010, Auckland, New Zealand
| | - Klaus Lehnert
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Euphemia Leung
- Auckland Cancer Society Research Centre, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Emily R Mears
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Russell Snell
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Richard Steele
- Department of Virology and Immunology, Auckland Hospital, Park Rd, Grafton, 1010, Auckland, New Zealand
| | - See-Tarn Woon
- Department of Virology and Immunology, Auckland Hospital, Park Rd, Grafton, 1010, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
50
|
Sideratou CM, Papaneophytou C. Persistent Vascular Complications in Long COVID: The Role of ACE2 Deactivation, Microclots, and Uniform Fibrosis. Infect Dis Rep 2024; 16:561-571. [PMID: 39051242 PMCID: PMC11270324 DOI: 10.3390/idr16040042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2), a key regulator in vasoregulation and the renin-angiotensin system, is hypothesized to be downregulated in patients with COVID-19, leading to a cascade of cardiovascular complications. This deactivation potentially results in increased blood pressure and vessel injury, contributing to the formation and persistence of microclots in the circulation. Herein, we propose a hypothesis regarding the prolonged vascular complications observed in long COVID, focusing on the role of ACE2 deactivation and/or shedding, the persistence of microclots, and the unique pattern of fibrosis induced by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). Furthermore, we propose that the distinctive, uniform fibrosis associated with COVID-19, which is challenging to detect through conventional X-ray imaging, exacerbates vascular injury and impairs oxygenation. The persistence of these microclots and the unique fibrosis pattern are suggested as key factors in the extended duration of vascular complications post-COVID-19 infection, regardless of the initial disease severity. Moreover, plasma ACE2 activity has the potential to serve as prognostic or diagnostic biomarkers for monitoring disease severity and managing long COVID symptoms. Elucidating the role of ACE2 deactivation and the consequent events is vital for understanding the long-term effects of COVID-19. The experimental verification of this hypothesis through in vitro studies, clinical longitudinal studies, and advanced imaging techniques could yield significant insights into the pathophysiological mechanisms underlying long COVID, thereby improving the management of patients, particularly those with cardiovascular complications.
Collapse
Affiliation(s)
| | - Christos Papaneophytou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus;
| |
Collapse
|