1
|
Camelo S, Dioh W, Teixeira JP, Busse LW, Nair G, Plantefeve G, Morelot-Panzini C, Lobo SM, Self WH, Collins SP, Van Maanen R, Veillet S. Modulation of the renin-angiotensin system against COVID-19: A path forward? Int J Infect Dis 2025; 154:107867. [PMID: 40049397 DOI: 10.1016/j.ijid.2025.107867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Soon after the pandemic outbreak in 2020, it was proposed that binding of SARS-CoV-2 to the angiotensin converting enzyme-2 may explain most of COVID-19's manifestations. Therefore, manipulation of the renin-angiotensin system (RAS) by using well known and commercialized blockers of its classical arm or by repurposing new stimulators of the alternative RAS pathway in clinical development was seen as a potentially effective strategy for the treatment of COVID-19. Moreover, this therapeutic approach had previously shown significant promise in the treatment of other respiratory viral respiratory infections and forms of acute respiratory distress syndrome. Consequently, several randomized clinical trials (RCTs) were launched to test the efficacy of rebalancing the RAS to reduce the severity of COVID-19. While most of these trials produced neutral results, certain studies reached their primary endpoints. In the present collaborative review, sponsors and main investigators of some of these trials attempt to reach a consensus regarding their clinical significance and which factors influenced their differing outcomes. The knowledge gained through the careful analysis of these RCTs of RAS modulators in patients with severe COVID-19 may prove useful for other forms of acute lung injury.
Collapse
Affiliation(s)
| | - Waly Dioh
- Biophytis, Sorbonne Université, Paris, France
| | - J Pedro Teixeira
- Divisions of Nephrology and Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of New Mexico School of Medicine MSC10-5550, 1 University of New Mexico, Albuquerque, New Mexico, USA
| | - Laurence W Busse
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Girish Nair
- OUWB School of Medicine, William Beaumont Hospital, Royal Oak, Michigan, USA
| | | | - Capucine Morelot-Panzini
- Sorbonne Université, INSERM, UMRS1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France; AP-HP, Groupe Hospitalier Universitaire APHP-Sorbonne Université, hôpital Pitié-Salpêtrière, Service de Pneumologie (Département R3S), Paris, France
| | - Suzana Margareth Lobo
- Intensive Care Division, Hospital de Base, Faculdade de Medicina de São José do Rio Preto - São José do Rio Preto (Sao Paulo), Brazil
| | - Wesley H Self
- Department of Emergency Medicine and Vanderbilt University Medical Center Veterans Affairs Tennessee Valley Healthcare System, Geriatric Research, Education and Clinical Center (GRECC), Nashville, Tennessee, USA
| | - Sean P Collins
- Department of Emergency Medicine and Vanderbilt University Medical Center Veterans Affairs Tennessee Valley Healthcare System, Geriatric Research, Education and Clinical Center (GRECC), Nashville, Tennessee, USA
| | | | | |
Collapse
|
2
|
Srivastava R, Panda SK, Sen Gupta PS, Chaudhary A, Naaz F, Yadav AK, Ram NK, Rana MK, Singh RK, Srivastava R. In silico evaluation of S-adenosyl-L-homocysteine analogs as inhibitors of nsp14-viral cap N7 methyltranferase and PLpro of SARS-CoV-2: synthesis, molecular docking, physicochemical data, ADMET and molecular dynamics simulations studies. J Biomol Struct Dyn 2025; 43:3258-3275. [PMID: 38147408 DOI: 10.1080/07391102.2023.2297005] [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: 07/18/2023] [Accepted: 12/13/2023] [Indexed: 12/28/2023]
Abstract
A series of S-adenosyl-L-homosysteine (SAH) analogs, with modification in the base and sugar moiety, have been designed, synthesized and screened as nsp14 and PLpro inhibitors of severe acute respiratory syndrome corona virus (SARS-CoV-2). The outcomes of ADMET (Adsorption, Distribution, Metabolism, Excretion, and Toxicity) studies demonstrated that the physicochemical properties of all analogs were permissible for development of these SAH analogs as antiviral agents. All molecules were screened against different SARS-CoV-2 targets using molecular docking. The docking results revealed that the SAH analogs interacted well in the active site of nsp14 protein having H-bond interactions with the amino acid residues Arg289, Val290, Asn388, Arg400, Phe401 and π-alkyl interactions with Arg289, Val290 and Phe426 of Nsp14-MTase site. These analogs also formed stable H-bonds with Leu163, Asp165, Arg167, Ser246, Gln270, Tyr274 and Asp303 residues of PLpro proteins and found to be quite stable complexes therefore behaved as probable nsp14 and PLpro inhibitors. Interestingly, analog 3 showed significant in silico activity against the nsp14 N7 methyltransferase of SARS-CoV-2. The molecular dynamics (MD) and post-MD results of analog 3 unambiguously established the higher stability of the nsp14 (N7 MTase):3 complex and also indicated its behavior as probable nsp14 inhibitor like the reference sinefungin. The docking and MD simulations studies also suggested that sinefungin did act as SARS-CoV-2 PLpro inhibitor as well. This study's findings not only underscore the efficacy of the designed SAH analogs as potent inhibitors against crucial SARS-CoV-2 proteins but also pinpoint analog 3 as a particularly promising candidate. All the study provides valuable insights, paving the way for potential advancements in antiviral drug development against SARS-CoV-2.
Collapse
Affiliation(s)
- Ritika Srivastava
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur, Odisha, India
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Allahabad, India
| | - Saroj Kumar Panda
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur, Odisha, India
| | - Parth Sarthi Sen Gupta
- School of Biosciences and Bioengineering, D Y Patil International University, Akurdi, India
| | - Anvita Chaudhary
- Department of Applied Chemistry, Delhi Technological University, Delhi, India
| | - Farha Naaz
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Allahabad, India
| | - Aditya K Yadav
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Allahabad, India
| | - Nand Kumar Ram
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Allahabad, India
| | - Malay Kumar Rana
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur, Odisha, India
| | - Ramendra K Singh
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Allahabad, India
| | - Richa Srivastava
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Allahabad, India
- Department of Applied Chemistry, Delhi Technological University, Delhi, India
| |
Collapse
|
3
|
Mukhija S, Sunog M, Magdamo C, Albers MW. Impact of Severe COVID-19 on Accelerating Dementia Onset: Clinical and Epidemiological Insights. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.26.25324688. [PMID: 40196257 PMCID: PMC11974976 DOI: 10.1101/2025.03.26.25324688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Importance Severe COVID-19 infection has been associated with neurological complications, but its role in accelerating cognitive decline remains unclear. Objective To determine whether individuals hospitalized for severe COVID-19 exhibit a higher incidence of new onset cognitive impairment compared to those hospitalized for other conditions. Design A retrospective study emulating a target trial using Mass General Brigham electronic health records (March 2020-August 2024). The causal effect of COVID-19 hospitalization was estimated via cumulative incidence functions accounting for the competing risk of death. Setting Multicenter hospital-based study across the Mass General Brigham healthcare system. Participants A total of 221613 hospitalized patients met the eligibility criteria, including 6454 (2.0%) admitted due to COVID-19 and 215159 (98.0%) for all other conditions. Patients were excluded if they had less than three months of follow-up (due to censoring, cognitive impairment, or death), were younger than 55 years at baseline, or had no prior visit to Mass General Brigham in the year before baseline. Main Outcomes and Measures The primary outcome was new-onset cognitive impairment, identified via ICD codes and dementia medication prescriptions. The primary analysis estimated the hazard ratio for cognitive impairment with COVID-19 hospitalization relative to other hospitalizations, along with the risk difference at 4.5 years estimated via cumulative incidence functions. Inverse propensity score weighting was used to balance covariates (age, sex, comorbidities, hospitalization period). Results Among eligible patients (mean [SD] age, 69.55 [9.42] years, 55% female), those hospitalized for COVID-19 were significantly older and had more comorbidities (p < 0.05). COVID-19 hospitalization was associated with a higher risk of developing cognitive impairment (Hazard Ratio: 1.14 [95% CI: 1.02-1.30], P = 0.018). At 4.5 years, the cumulative incidence of cognitive impairment was 12.5% [95% CI: 11.3-13.5] in the COVID-19 group, compared to 11.6% [95% CI: 11.1-12.1] in the non-COVID-19 group. Conclusions and Relevance Severe COVID-19 infection was associated with an elevated risk of developing clinically recognized cognitive impairment. Future studies are needed to validate findings in other health care settings. Early screening and intervention for cognitive decline may help optimize long-term outcomes for COVID-19 patients.
Collapse
Affiliation(s)
- Sasha Mukhija
- Neurology Department, Massachusetts General Hospital, Boston, USA (Mukhija, Sunog, Magdamo, Albers)
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, USA (Mukhija, Magdamo, Albers)
- Universitätsspital Zürich, Zürich, Switzerland (Mukhija)
| | - Max Sunog
- Neurology Department, Massachusetts General Hospital, Boston, USA (Mukhija, Sunog, Magdamo, Albers)
| | - Colin Magdamo
- Neurology Department, Massachusetts General Hospital, Boston, USA (Mukhija, Sunog, Magdamo, Albers)
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, USA (Mukhija, Magdamo, Albers)
| | - Mark W. Albers
- Neurology Department, Massachusetts General Hospital, Boston, USA (Mukhija, Sunog, Magdamo, Albers)
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, USA (Mukhija, Magdamo, Albers)
| |
Collapse
|
4
|
Mokra D, Porvaznik I, Mokry J. N-Acetylcysteine in the Treatment of Acute Lung Injury: Perspectives and Limitations. Int J Mol Sci 2025; 26:2657. [PMID: 40141299 PMCID: PMC11942046 DOI: 10.3390/ijms26062657] [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: 02/05/2025] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
N-acetylcysteine (NAC) can take part in the treatment of chronic respiratory diseases because of the potent mucolytic, antioxidant, and anti-inflammatory effects of NAC. However, less is known about its use in the treatment of acute lung injury. Nowadays, an increasing number of studies indicates that early administration of NAC may reduce markers of oxidative stress and alleviate inflammation in animal models of acute lung injury (ALI) and in patients suffering from distinct forms of acute respiratory distress syndrome (ARDS) or pulmonary infections including community-acquired pneumonia or Coronavirus Disease (COVID)-19. Besides low costs, easy accessibility, low toxicity, and rare side effects, NAC can also be combined with other drugs. This article provides a review of knowledge on the mechanisms of inflammation and oxidative stress in various forms of ALI/ARDS and critically discusses experience with the use of NAC in these disorders. For preparing the review, articles published in the English language from the PubMed database were used.
Collapse
Affiliation(s)
- Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia
| | - Igor Porvaznik
- Department of Laboratory Medicine, Faculty of Health Sciences, Catholic University in Ružomberok, SK-03401 Ružomberok, Slovakia;
| | - Juraj Mokry
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia;
| |
Collapse
|
5
|
Adilović M. COVID-19 related complications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:259-314. [PMID: 40246346 DOI: 10.1016/bs.pmbts.2025.02.002] [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 COVID-19 pandemic has significantly impacted global healthcare systems, revealed vulnerabilities and prompted a re-evaluation of medical practices. Acute complications from the virus, including cardiovascular and neurological issues, have underscored the necessity for timely medical interventions. Advances in diagnostic methods and personalized therapies have been pivotal in mitigating severe outcomes. Additionally, Long COVID has emerged as a complex challenge, affecting various body systems and leading to respiratory, cardiovascular, neurological, psychological, and musculoskeletal problems. This broad spectrum of complications highlights the importance of multidisciplinary management approaches that prioritize therapy, rehabilitation, and patient-centered care. Vulnerable populations such as paediatric patients, pregnant women, and immunocompromised individuals face unique risks and complications, necessitating continuous monitoring and tailored management strategies to reduce morbidity and mortality associated with COVID-19.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnička cesta, Sarajevo, Bosnia and Herzegovina.
| |
Collapse
|
6
|
Chen Y, Jan J, Yang C, Yen T, Linh TTD, Annavajjula S, Satapathy MK, Tsao S, Hsieh C. Cognitive Sequelae of COVID-19: Mechanistic Insights and Therapeutic Approaches. CNS Neurosci Ther 2025; 31:e70348. [PMID: 40152069 PMCID: PMC11950837 DOI: 10.1111/cns.70348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic has left an indelible mark on the world, with mounting evidence suggesting that it not only posed acute challenges to global healthcare systems but has also unveiled a complex array of long-term consequences, particularly cognitive impairment (CI). As the persistence of post-COVID-19 neurological syndrome could evolve into the next public health crisis, it is imperative to gain a better understanding of the intricate pathophysiology of CI in COVID-19 patients and viable treatment strategies. METHODS This comprehensive review explores the pathophysiology and management of cognitive impairment across the phases of COVID-19, from acute infection to Long-COVID, by synthesizing findings from clinical, preclinical, and mechanistic studies to identify key contributors to CI, as well as current therapeutic approaches. RESULTS Key mechanisms contributing to CI include persistent neuroinflammation, cerebrovascular complications, direct neuronal injury, activation of the kynurenine pathway, and psychological distress. Both pharmacological interventions, such as anti-inflammatory therapies and agents targeting neuroinflammatory pathways, and non-pharmacological strategies, including cognitive rehabilitation, show promise in addressing these challenges. Although much of the current evidence is derived from preclinical and animal studies, these findings provide foundational insights into potential treatment approaches. CONCLUSION By synthesizing current knowledge, this review highlights the importance of addressing COVID-19-related cognitive impairment and offers actionable insights for mitigation and recovery as the global community continues to grapple with the pandemic's long-term impact.
Collapse
Affiliation(s)
- Yu‐Hao Chen
- Section of Neurosurgery, Department of SurgeryDitmanson Medical Foundation, Chia‐Yi Christian HospitalChia‐Yi CityTaiwan
- Chung‐Jen Junior College of Nursing, Health Sciences and ManagementChia‐Yi CountryTaiwan
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Jing‐Shiun Jan
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Chih‐Hao Yang
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Ting‐Lin Yen
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
- Department of Medical ResearchCathay General HospitalTaipeiTaiwan
| | - Tran Thanh Duy Linh
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
- Family Medicine Training Center, University of Medicine and Pharmacy at Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Saileela Annavajjula
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Mantosh Kumar Satapathy
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Shin‐Yi Tsao
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
- Division of Endocrinology and Metabolism, Department of Internal MedicineTaipeiTaiwan
| | - Cheng‐Ying Hsieh
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
7
|
Chun KH, Kim HJ, Kang DR, Kim JY, Kim W, Jeong YW, Han SH, Koh KK. Sex-specific impact of the COVID-19 outbreak on the incidence of metabolic syndrome: a comparative study of 2018-2019 and 2020-2021. Korean J Intern Med 2025; 40:262-274. [PMID: 40102710 PMCID: PMC11938662 DOI: 10.3904/kjim.2024.288] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 10/15/2024] [Accepted: 11/20/2024] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND/AIMS The coronavirus disease 2019 (COVID-19) pandemic has significantly impacted global health, exacerbated metabolic health issues, and altered lifestyle behaviors. This study examined the sex-specific impact of the COVID-19 outbreak on the incidence of metabolic syndrome using data from the Korea National Health and Nutrition Examination Survey (KNHANES). METHODS Data from the KNHANES VII (2018) and VIII (2019-2021), including 15,499 participants, were analyzed. The study population was stratified by sex, and further subdivisions were conducted based on the timeframe relative to the COVID-19 outbreak. Variables such as age, education level, household income, smoking status, and high-risk drinking were analyzed to assess their influence on the prevalence of metabolic syndrome. RESULTS The overall prevalence of metabolic syndrome significantly increased from 28.11% before the outbreak to 29.69% after the outbreak. Both males and females reported significant increases in waist circumference and fasting glucose levels. Age and education level differentially influenced the prevalence of metabolic syndrome between the sex. Smoking was significantly associated with increased prevalence in males, whereas high-risk drinking was associated with increased prevalence in males and decreased prevalence in females. CONCLUSION The COVID-19 pandemic has significantly increased the prevalence of metabolic syndrome with notable sex-specific differences. These findings highlight the need for sex-specific public health interventions to mitigate the impact of the pandemic on metabolic health.
Collapse
Affiliation(s)
- Kyeong-Hyeon Chun
- Division of Cardiology, National Health Insurance Service Ilsan Hospital, Goyang,
Korea
| | - Hyun-Jin Kim
- Division of Cardiology, Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri,
Korea
| | - Dae Ryong Kang
- Department of Precision Medicine, Yonsei University Wonju College of Medicine, Wonju,
Korea
| | - Jang Young Kim
- Division of Cardiology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju,
Korea
| | - Wonjin Kim
- Division of Endocrinology, Department of Internal Medicine, CHA University School of Medicine, Pocheon,
Korea
| | - Yong Whi Jeong
- Department of Medical Informatics and Biostatistics, Graduate School, Yonsei University, Wonju,
Korea
| | - Seung Hwan Han
- Division of Cardiology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon,
Korea
| | - Kwang Kon Koh
- Division of Cardiology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon,
Korea
| |
Collapse
|
8
|
Pencheva M, Manchorova-Veleva N, Baruh D, Rusinov G, Vangelov L. Analysis of Biomarker Levels in Nasopharyngeal Swabs, Serum, and Saliva Across Different Health Conditions. Life (Basel) 2025; 15:324. [PMID: 40003732 PMCID: PMC11857456 DOI: 10.3390/life15020324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/04/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is associated with a wide variety of clinical manifestations. AIM This study aims to evaluate the levels of angiotensin-converting enzyme 2 (ACE2), metalloprotease 17 (ADAM17), Interleukin-17A (IL-17A), transmembrane serine protease 2 (TMPRSS2), apelin (AP), and vitamin D (VD) biomarkers in nasopharyngeal swab (NPS), serum, and saliva, as well as the change in their values depending on the health status of individuals. MATERIAL AND METHODS The analysis was performed by using enzyme-linked immunosorbent assay (ELISA) methods. RESULTS Comparing the levels of the investigated markers in saliva, we found significantly elevated ACE2 values in vaccinated patients, followed by those with severe COVID-19, compared to healthy, previously infected, and mild COVID-19 groups. For TMPRSS2, IL-17A, ADAM-17, and AP, values were significantly higher in all non-healthy groups (previously infected, mild, and severe COVID-19) compared to healthy individuals. Serum levels of VD were consistently low across all five studied groups, suggesting values below normal ranges. Analysis of marker data in saliva, NPS, and serum revealed a positive correlation between NPS and serum and saliva and serum, as well as between saliva and NPS for all studied markers. CONCLUSIONS In summary, monitoring changes in biomarkers present in Saliva holds promise as a predictive tool for various diseases. This approach enables the early implementation of preventive measures and protective strategies, potentially improving overall health outcomes.
Collapse
Affiliation(s)
- Mina Pencheva
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Neshka Manchorova-Veleva
- Department of Operative Dentistry and Endodontics, Faculty of Dental Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (N.M.-V.); (L.V.)
| | - David Baruh
- Department of Software Engineering, Faculty of Mathematics and Informatics, Sofia University “St. Kliment Ohridski”, 1164 Sofia, Bulgaria;
| | - Georgi Rusinov
- Clinic of Infectious Diseases, University Hospital St. George JSC in Plovdiv, 4021 Plovdiv, Bulgaria;
| | - Lyubomir Vangelov
- Department of Operative Dentistry and Endodontics, Faculty of Dental Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (N.M.-V.); (L.V.)
| |
Collapse
|
9
|
Eltayeb A, Redwan EM. T-cell immunobiology and cytokine storm of COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:1-30. [PMID: 40246342 DOI: 10.1016/bs.pmbts.2024.10.003] [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 2019 coronavirus illness (COVID 2019) first manifests as a newly identified pneumonia and may quickly escalate to acute respiratory distress syndrome, which has caused a global pandemic. Except for individualized supportive care, no curative therapy has been steadfastly advised for COVID-19 up until this point. T cells and virus-specific T lymphocytes are required to guard against viral infection, particularly COVID-19. Delayed immunological reconstitution (IR) and cytokine storm (CS) continue to be significant barriers to COVID-19 cure. While severe COVID-19 patients who survived the disease had considerable lymphopenia and increased neutrophils, especially in the elderly, their T cell numbers gradually recovered. Exhausted T lymphocytes and elevated levels of pro-inflammatory cytokines, including IL6, IL10, IL2, and IL17, are observed in peripheral blood and the lungs. It implies that while convalescent plasma, IL-6 blocking, mesenchymal stem cells, and corticosteroids might decrease CS, Thymosin α1 and adaptive COVID-19-specific T cells could enhance IR. There is an urgent need for more clinical research in this area throughout the world to open the door to COVID-19 treatment in the future.
Collapse
Affiliation(s)
- Ahmed Eltayeb
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
10
|
Lissoni P, Rovelli F, Messina G, Monzon A, Valentini A, Sassola A, Di Fede G, Simoes-de-Silva AC, Merli N, Bartsch C, Vlaescu VG, Cardinali DD. Psycho-Neuro-EndocrinE-Immunology Therapy of Cancer, Autoimmunity, Geriatric Disorders, Covid-19, and Hypertension. Methods Mol Biol 2025; 2868:111-132. [PMID: 39546228 DOI: 10.1007/978-1-0716-4200-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Despite the great number of experimental investigations in the area of psycho-neuro-endocrine-immunology showing that endocrine, nervous, and immune systems cannot be in vivo physiologically separated, the diagnosis and therapies of the pathologies of these three functional biological systems continue to be separately performed from a clinical practice point of view. The separation between experimental and clinical medicine became dramatic after the discovery of more than 10 human molecules provided by anti-inflammatory and antitumor activity, completely devoid of any toxicity, which may be subdivided into three fundamental classes, consisting of the pineal indole, beta-carboline, and methoxy-kynuramine hormones. Moreover, human systemic diseases, including cancer, autoimmunity, and cardiovascular pathologies, despite their different pathogenesis and symptomatology, are commonly characterized by a progressive decline in the endogenous production of pineal hormones, endocannabinoids, and Ang 1-7, with a consequent inflammatory status and diminished natural resistance against cancer. Then the evaluation of the functional status of the pineal gland, the endocannabinoid system, and ACE2-Ang 1-7 axis should have to be included within the laboratory analyses for the systemic diseases. Finally, the correction of cancer- and autoimmunity-related neuroimmune and neuroendocrine alterations could influence the clinical course of systemic diseases. In fact, preliminary clinical results would demonstrate that the neuroimmune regimen with pineal hormones, cannabinoids, and Ang 1-7 may allow clinical benefits also in patients affected by systemic pathologies, including cancer, autoimmunity, and cardiovascular diseases, who did not respond to the standard therapies.
Collapse
Affiliation(s)
- Paolo Lissoni
- Institute of Biological Medicine, Milan, Italy
- Madonna del Soccorso Hospital, San Benedetto del Tronto, Ascoli Piceno, Italy
| | - Franco Rovelli
- Institute of Biological Medicine, Milan, Italy
- Madonna del Soccorso Hospital, San Benedetto del Tronto, Ascoli Piceno, Italy
| | - Giusy Messina
- Institute of Biological Medicine, Milan, Italy
- Madonna del Soccorso Hospital, San Benedetto del Tronto, Ascoli Piceno, Italy
| | - Alejandra Monzon
- Institute of Biological Medicine, Milan, Italy
- Madonna del Soccorso Hospital, San Benedetto del Tronto, Ascoli Piceno, Italy
| | - Agnese Valentini
- Institute of Biological Medicine, Milan, Italy
- Madonna del Soccorso Hospital, San Benedetto del Tronto, Ascoli Piceno, Italy
| | | | | | | | | | | | | | | |
Collapse
|
11
|
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
|
12
|
Narayanan SN, Padiyath S, Chandrababu K, Raj L, P S BC, Ninan GA, Sivadasan A, Jacobs AR, Li YW, Bhaskar A. Neurological, psychological, psychosocial complications of long-COVID and their management. Neurol Sci 2025; 46:1-23. [PMID: 39516425 PMCID: PMC11698801 DOI: 10.1007/s10072-024-07854-5] [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: 09/13/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Since it first appeared, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has had a significant and lasting negative impact on the health and economies of millions of individuals all over the globe. At the level of individual health too, many patients are not recovering fully and experiencing a long-term condition now commonly termed 'long-COVID'. Long-COVID is a collection of symptoms which must last more than 12 weeks following initial COVID infection, and which cannot be adequately explained by alternate diagnoses. The neurological and psychosocial impact of long-COVID is itself now a global health crisis and therefore preventing, diagnosing, and managing these patients is of paramount importance. This review focuses primarily on: neurological functioning deficits; mental health impacts; long-term mood problems; and associated psychosocial issues, among patients suffering from long-COVID with an eye towards the neurological basis of these symptoms. A concise account of the clinical relevance of the neurological and psychosocial impacts of long-COVID, the effects on long-term morbidity, and varied approaches in managing patients with significant chronic neurological symptoms and conditions was extracted from the literature, analysed and reported. A comprehensive account of plausible pathophysiological mechanisms involved in the development of long-COVID, its management, and future research needs have been discussed.
Collapse
Affiliation(s)
- Sareesh Naduvil Narayanan
- Department of Physiology, School of Medicine and Dentistry, AUC-UK Track, University of Central Lancashire, Preston, UK.
| | - Sreeshma Padiyath
- Department of Microbiology, School of Medicine and Dentistry, AUC-UK Track, University of Central Lancashire, Preston, UK
| | - Krishnapriya Chandrababu
- Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology (CUSAT), Kochi, India
| | - Lima Raj
- Department of Psychology, Sree Sankaracharya University of Sanskrit, Kalady, India
| | - Baby Chakrapani P S
- Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology (CUSAT), Kochi, India
- Centre for Excellence in Neurodegeneration and Brain Health (CENABH), Cochin University of Science and Technology (CUSAT), Kochi, India
| | | | - Ajith Sivadasan
- Department of Neurology, Christian Medical College (CMC), Vellore, India
| | - Alexander Ryan Jacobs
- School of Medicine and Dentistry, AUC-UK Track, University of Central Lancashire, Preston, UK
| | - Yan Wa Li
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Anand Bhaskar
- Department of Physiology, Christian Medical College (CMC), Vellore, India
| |
Collapse
|
13
|
Guerrerio AL, Mateja A, MacCarrick G, Fintzi J, Brittain E, Frischmeyer-Guerrerio PA, Dietz HC. Cardiovascular complications in vascular connective tissue disorders after COVID-19 infection and vaccination. PLoS One 2024; 19:e0315499. [PMID: 39705273 DOI: 10.1371/journal.pone.0315499] [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: 05/17/2024] [Accepted: 11/26/2024] [Indexed: 12/22/2024] Open
Abstract
BACKGROUND COVID-19 infection and vaccination have been reported to confer an elevated risk for cardiovascular events (CVE). We sought to determine whether individuals with an underlying vascular connective tissue disorder including Marfan syndrome (MFS), Loeys-Dietz syndrome (LDS), or vascular Ehlers Danlos syndrome (vEDS) are at increased risk for cardiac events after COVID-19 infection or vaccination. METHODS 325 respondents self-reported data through a cross-sectional, web-based survey available from 22 November 2021, through 15 March 2022 regarding COVID-19 illness and vaccinations, the occurrence of any CVE, and adverse events following vaccination. The data were analyzed using a Cox proportional hazards model with time varying indicators for COVID-19 illness/vaccination in the preceding 30 days. RESULTS COVID-19 illness was significantly associated with an increased rate of a new abnormal heart rhythm 30 days following infection. No other CVEs were reported in the 90 days after COVID-19 illness. We did not find evidence of an increased rate of any CVE in the 30 days following any COVID-19 vaccination dose. CONCLUSION In respondents with MFS, LDS, or vEDS, we uncovered no evidence of an increase in CVEs in the 30 days following COVID-19 illness, with the possible exception of dysrhythmia. In light of the absence of a substantial increase in self-reported CVEs in the 30 days following COVID-19 vaccination, these data are in keeping with the recommendation from the Marfan Foundation Professional Advisory Board that all eligible persons be vaccinated for COVID-19.
Collapse
Affiliation(s)
- Anthony L Guerrerio
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Allyson Mateja
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research. Frederick, Maryland, United States of America
| | - Gretchen MacCarrick
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jonathan Fintzi
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Erica Brittain
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Pamela A Frischmeyer-Guerrerio
- The Laboratory of Allergic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Harry C Dietz
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| |
Collapse
|
14
|
Uddin MN, Mia MA, Akter Y, Chowdhury MAB, Rahman MH, Siddiqua H, Shathi US, Al-Mamun A, Siddika F, Marzan LW. Variations in Furin SNPs, a Major Concern of SARS-CoV-2 Susceptibility Among Different Populations: An In- Silico Approach. Bioinform Biol Insights 2024; 18:11779322241306388. [PMID: 39703750 PMCID: PMC11656424 DOI: 10.1177/11779322241306388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
COVID-19 caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) had an adverse effect globally because it caused a global pandemic with several million deaths. This virus possesses spike protein that is cleaved or activated by Furin-like protease enzymes occurring by mammalian lung or respiratory cells to enter the mammalian body. The addition of the Furin cleavage site in SARS-CoV-2 makes it a more infectious and emerging virus than its ancestor's viruses. Phylogenetic relationships of coronavirus spike proteins have analyzed and mapped Furin recognition motif on the tree using bioinformatics tools such as GTEx, KEGG, GO, NCBI, PolyPhen-2, SNAP2, PANTHER, Hidden Markov Models (Fathmm), Phd-single-nucleotide polymorphism (SNP), I-TASSER, Modpred, Phobius, SIFT, iPTREE-STAB, and PROVEAN. During this study, it has been found that in certain regions, Furin SNPs have some relation with the susceptibility to SARS-CoV-2. Whereas in other regions, the effects are very negligible. Finally, our study demonstrates that Furin SNPs have a strong relationship with susceptibility to SARS-CoV-2. As it helps to cleave the spike protein of the virus, thus it can be targeted to inhibit at a particular site to prevent the SARS-CoV-2 from the entrance into the body.
Collapse
Affiliation(s)
- Md Nasir Uddin
- Laboratory of Microbial Genomics and Metabolic Engineering, Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Md Arzo Mia
- Laboratory of Microbial Genomics and Metabolic Engineering, Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Yasmin Akter
- Laboratory of Microbial Genomics and Metabolic Engineering, Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Mohammad Al-baruni Chowdhury
- Laboratory of Microbial Genomics and Metabolic Engineering, Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Md Hadisur Rahman
- Molecular Biotechnology Division, National Institute of Biotechnology, Savar, Bangladesh
| | - Hafsa Siddiqua
- Laboratory of Microbial Genomics and Metabolic Engineering, Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Umme Salma Shathi
- Laboratory of Microbial Genomics and Metabolic Engineering, Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Abdullah Al-Mamun
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, Bangladesh
| | - Farida Siddika
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Lolo Wal Marzan
- Laboratory of Microbial Genomics and Metabolic Engineering, Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| |
Collapse
|
15
|
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
|
16
|
Dos Santos L, Favaroni Mendes Salgado Ribeiro LA, Febba Gomes AC, Azinheira Nobrega Cruz N, Gonçalves de Oliveira LC, Cenedeze MA, Tedesco Silva Junior H, Medina Pestana JO, Casarini DE. ACE and ACE2 activities and polymorphisms assessment: A populational study from Ipaussu (SP, Brazil) during the COVID-19 pandemic. Life Sci 2024; 358:123157. [PMID: 39437850 DOI: 10.1016/j.lfs.2024.123157] [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: 05/21/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
AIM The angiotensin-converting enzyme 2 (ACE2) and its homolog, the angiotensin converting enzyme 1 (ACE), are involved in COVID-19 physiopathology. Alterations in the enzymatic structure, expression, and/or activity may influence the risk of infection and severity of disease. For this reason, we aimed to identify different allelic forms of ACE2 G8790A and ACE I/D polymorphisms in a Brazilian cohort and evaluate their impact on ACE and ACE2 activities and their association with COVID-19 susceptibility and severity. MAIN METHODS A total of 549 COVID-19-negative and 270 COVID-19-positive participants from Ipaussu, Sao Paulo, Brazil, were recruited. ACE2 and ACE activities were measured by fluorogenic assays using MCA-Ala-Pro-Lys(Dnp) as the substrate for ACE2 and Z-Phe-His-Leu-OH (Z-FHL) and Hippuryl-His-Leu-OH (h-HL) as substrates for ACE. Genomic DNA was extracted from EDTA-peripheral blood, and the regions of the genes containing ACE2 G8790A and ACE I/D polymorphisms were amplified by PCR-restriction fragment length polymorphism and real-time PCR, respectively. KEY FINDINGS The G allele of ACE2 G8790A polymorphism and D allele of ACE I/D polymorphism are associated with increased ACE and ACE2 activities. ACE activity ratio (Z-FHL/h-HL), an inflammatory marker, is increased in women with GG genotype and COVID-19-positive diagnosis. SIGNIFICANCE For the first time, it was demonstrated that in females, the GG genotype is associated with increased ACE activity ratio (Z-FHL/h-HL) in the COVID-19-positive group. Elevated ACE activity ratio (Z-FHL/h-HL) is highly linked to inflammation and may justify the associations between the G genotype and COVID-19 severity of symptoms and outcomes.
Collapse
Affiliation(s)
- Lilian Dos Santos
- Department of Medicine, Discipline of Nephrology, University Federal of Sao Paulo, Sao Paulo, Brazil
| | | | | | | | | | - Marcos Antonio Cenedeze
- Department of Medicine, Discipline of Nephrology, University Federal of Sao Paulo, Sao Paulo, Brazil
| | | | - José Osmar Medina Pestana
- Department of Medicine, Discipline of Nephrology, University Federal of Sao Paulo, Sao Paulo, Brazil
| | - Dulce Elena Casarini
- Department of Medicine, Discipline of Nephrology, University Federal of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
17
|
Yu Y, Li GF, Li J, Han LY, Zhang ZL, Liu TS, Jiao SX, Qiao YW, Zhang N, Zhan DC, Tang SQ, Yu G. Ursodeoxycholic acid and COVID-19 outcomes: a cohort study and data synthesis of state-of-art evidence. Expert Rev Anti Infect Ther 2024; 22:1239-1250. [PMID: 38975666 DOI: 10.1080/14787210.2024.2376153] [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: 03/26/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND The potential of ursodeoxycholic acid (UDCA) in inhibiting angiotensin-converting enzyme 2 was demonstrated. However, conflicting evidence emerged regarding the association between UDCA and COVID-19 outcomes, prompting the need for a comprehensive investigation. RESEARCH DESIGN AND METHODS Patients diagnosed with COVID-19 infection were retrospectively analyzed and divided into two groups: the UDCA-treated group and the control group. Kaplan-Meier recovery analysis and Cox proportional hazards models were used to evaluate the recovery time and hazard ratios. Additionally, study-level pooled analyses for multiple clinical outcomes were performed. RESULTS In the 115-patient cohort, UDCA treatment was significantly associated with a reduced recovery time. The subgroup analysis suggests that the 300 mg subgroup had a significant (adjusted hazard ratio: 1.63 [95% CI, 1.01 to 2.60]) benefit with a shorter duration of fever. The results of pooled analyses also show that UDCA treatment can significantly reduce the incidence of severe/critical diseases in COVID-19 (adjusted odds ratio: 0.68 [95% CI, 0.50 to 0.94]). CONCLUSIONS UDCA treatment notably improves the recovery time following an Omicron strain infection without observed safety concerns. These promising results advocate for UDCA as a viable treatment for COVID-19, paving the way for further large-scale and prospective research to explore the full potential of UDCA.
Collapse
Affiliation(s)
- Yang Yu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, China
- General Foundation Department, Polixir.ai, Nanjing, China
| | - Guo-Fu Li
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jian Li
- Hospital of Nanjing University, Nanjing University, Nanjing, China
| | - Lu-Yao Han
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhi-Long Zhang
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, China
- General Foundation Department, Polixir.ai, Nanjing, China
| | - Tian-Shuo Liu
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, China
- General Foundation Department, Polixir.ai, Nanjing, China
| | - Shu-Xin Jiao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yu-Wei Qiao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Na Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - De-Chuan Zhan
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, China
| | - Shao-Qiu Tang
- Hospital of Nanjing University, Nanjing University, Nanjing, China
| | - Guo Yu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
18
|
Pacnejer AM, Butuca A, Dobrea CM, Arseniu AM, Frum A, Gligor FG, Arseniu R, Vonica RC, Vonica-Tincu AL, Oancea C, Mogosan C, Popa Ilie IR, Morgovan C, Dehelean CA. Neuropsychiatric Burden of SARS-CoV-2: A Review of Its Physiopathology, Underlying Mechanisms, and Management Strategies. Viruses 2024; 16:1811. [PMID: 39772122 PMCID: PMC11680421 DOI: 10.3390/v16121811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
The COVID-19 outbreak, caused by the SARS-CoV-2 virus, was linked to significant neurological and psychiatric manifestations. This review examines the physiopathological mechanisms underlying these neuropsychiatric outcomes and discusses current management strategies. Primarily a respiratory disease, COVID-19 frequently leads to neurological issues, including cephalalgia and migraines, loss of sensory perception, cerebrovascular accidents, and neurological impairment such as encephalopathy. Lasting neuropsychological effects have also been recorded in individuals following SARS-CoV-2 infection. These include anxiety, depression, and cognitive dysfunction, suggesting a lasting impact on mental health. The neuroinvasive potential of the virus, inflammatory responses, and the role of angiotensin-converting enzyme 2 (ACE2) in neuroinflammation are critical factors in neuropsychiatric COVID-19 manifestations. In addition, the review highlights the importance of monitoring biomarkers to assess Central Nervous System (CNS) involvement. Management strategies for these neuropsychiatric conditions include supportive therapy, antiepileptic drugs, antithrombotic therapy, and psychotropic drugs, emphasizing the need for a multidisciplinary approach. Understanding the long-term neuropsychiatric implications of COVID-19 is essential for developing effective treatment protocols and improving patient outcomes.
Collapse
Affiliation(s)
- Aliteia-Maria Pacnejer
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania; (A.-M.P.); (C.A.D.)
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.M.D.); (A.M.A.); (A.F.); (F.G.G.); (R.C.V.); (A.L.V.-T.); (C.M.)
| | - Anca Butuca
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.M.D.); (A.M.A.); (A.F.); (F.G.G.); (R.C.V.); (A.L.V.-T.); (C.M.)
| | - Carmen Maximiliana Dobrea
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.M.D.); (A.M.A.); (A.F.); (F.G.G.); (R.C.V.); (A.L.V.-T.); (C.M.)
| | - Anca Maria Arseniu
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.M.D.); (A.M.A.); (A.F.); (F.G.G.); (R.C.V.); (A.L.V.-T.); (C.M.)
| | - Adina Frum
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.M.D.); (A.M.A.); (A.F.); (F.G.G.); (R.C.V.); (A.L.V.-T.); (C.M.)
| | - Felicia Gabriela Gligor
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.M.D.); (A.M.A.); (A.F.); (F.G.G.); (R.C.V.); (A.L.V.-T.); (C.M.)
| | - Rares Arseniu
- County Emergency Clinical Hospital “Pius Brînzeu”, 300723 Timișoara, Romania;
| | - Razvan Constantin Vonica
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.M.D.); (A.M.A.); (A.F.); (F.G.G.); (R.C.V.); (A.L.V.-T.); (C.M.)
| | - Andreea Loredana Vonica-Tincu
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.M.D.); (A.M.A.); (A.F.); (F.G.G.); (R.C.V.); (A.L.V.-T.); (C.M.)
| | - Cristian Oancea
- Department of Pulmonology, Center for Research and Innovation in Personalized Medicine of Respiratory Diseases, “Victor Babeş” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Cristina Mogosan
- Department of Pharmacology, Physiology and Pathophysiology, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400029 Cluj-Napoca, Romania;
| | - Ioana Rada Popa Ilie
- Department of Endocrinology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 3-5 Louis Pasteur Street, 400349 Cluj-Napoca, Romania;
| | - Claudiu Morgovan
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.M.D.); (A.M.A.); (A.F.); (F.G.G.); (R.C.V.); (A.L.V.-T.); (C.M.)
| | - Cristina Adriana Dehelean
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania; (A.-M.P.); (C.A.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania
| |
Collapse
|
19
|
Moulias A, Koros R, Papageorgiou A, Katechis S, Patrinos P, Trigka-Vasilakopoulou A, Papageorgiou A, Papaioannou O, Akinosoglou K, Leventopoulos G, Tsigkas G, Tzouvelekis A, Davlouros P. Assessment of Endothelial Function in Patients with COVID-19 Using Peripheral Arterial Tonometry. Life (Basel) 2024; 14:1512. [PMID: 39598310 PMCID: PMC11595729 DOI: 10.3390/life14111512] [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: 10/11/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
There is increasing evidence that COVID-19 induces endothelial dysfunction that may precede thrombotic and cardiovascular complications. The aim of this study is to evaluate endothelial function using peripheral arterial tonometry (EndoPAT). The primary endpoint is the hyperemic vascular response index (LnRHI) at two months post-discharge. Secondary endpoints include the LnRHI during hospitalization and at six-month follow-up, the proportion of patients with endothelial dysfunction (LnRHI ≤ 0.51), and the incidence of thrombotic events, cardiovascular complications, and mortality during the follow-up period. The study included 23 COVID-19 patients and 22 COVID-19-negative, matched controls. The patients exhibited a significant reduction in the LnRHI at two months post-discharge compared to the controls (median = 0.55 [IQR: 0.49-0.68] vs. median = 0.70 [IQR: 0.62-0.83]; p = 0.012). The difference in the LnRHI between patients and controls was evident from hospitalization and persisted at two and six months without significant temporal changes. The proportion of COVID-19 patients with endothelial dysfunction (LnRHI ≤ 0.51) was 61% during hospitalization and 55% at six months. There was no significant difference in thrombotic or cardiovascular events, nor in mortality. This study demonstrates that COVID-19 adversely affects endothelial function, as evidenced by a reduction in the hyperemic vascular response index, and endothelial dysfunction may also persist.
Collapse
Affiliation(s)
- Athanasios Moulias
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Rafail Koros
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Angeliki Papageorgiou
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
- Department of Inherited and Rare Cardiovascular Diseases, Onassis Cardiac Surgery Center, 17674 Athens, Greece
| | - Spyridon Katechis
- Department of Rheumatology, General Hospital of Asklipieio Voulas, 16673 Athens, Greece;
| | - Panagiotis Patrinos
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Aikaterini Trigka-Vasilakopoulou
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Athanasios Papageorgiou
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Ourania Papaioannou
- Department of Pneumonology, General University Hospital of Patras, 26504 Patras, Greece; (O.P.); (A.T.)
| | - Karolina Akinosoglou
- Department of Internal Medicine, General University Hospital of Patras, 26504 Patras, Greece;
| | - Georgios Leventopoulos
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Grigorios Tsigkas
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Argyrios Tzouvelekis
- Department of Pneumonology, General University Hospital of Patras, 26504 Patras, Greece; (O.P.); (A.T.)
| | - Periklis Davlouros
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| |
Collapse
|
20
|
Rahmani D, Jafari A, Kesharwani P, Sahebkar A. Molecular targets in SARS-CoV-2 infection: An update on repurposed drug candidates. Pathol Res Pract 2024; 263:155589. [PMID: 39276508 DOI: 10.1016/j.prp.2024.155589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
The 2019 widespread contagion of the human coronavirus novel type (SARS-CoV-2) led to a pandemic declaration by the World Health Organization. A daily increase in patient numbers has formed an urgent necessity to find suitable targets and treatment options for the novel coronavirus (COVID-19). Despite scientists' struggles to discover quick treatment solutions, few effective specific drugs are approved to control SARS-CoV-2 infections thoroughly. Drug repositioning or Drug repurposing and target-based approaches are promising strategies for facilitating the drug discovery process. Here, we review current in silico, in vitro, in vivo, and clinical updates regarding proposed drugs for prospective treatment options for COVID-19. Drug targets that can direct pharmaceutical sciences efforts to discover new drugs against SARS-CoV-2 are divided into two categories: Virus-based targets, for example, Spike glycoprotein and Nucleocapsid Protein, and host-based targets, for instance, inflammatory cytokines and cell receptors through which the virus infects the cell. A broad spectrum of drugs has been found to show anti-SARS-CoV-2 potential, including antiviral drugs and monoclonal antibodies, statins, anti-inflammatory agents, and herbal products.
Collapse
Affiliation(s)
- Dibachehr Rahmani
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ameneh Jafari
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
21
|
Khanal R, Heinen N, Bogomolova A, Meister TL, Herrmann ST, Westhoven S, Nocke MK, Todt D, Jockenhövel F, Klein IM, Hartmann L, Vondran FWR, Steinmann E, Zimmer G, Ott M, Brown RJP, Sharma AD, Pfaender S. MicroRNAs modulate SARS-CoV-2 infection of primary human hepatocytes by regulating the entry factors ACE2 and TMPRSS2. Liver Int 2024; 44:2983-2995. [PMID: 39175256 DOI: 10.1111/liv.16079] [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: 08/07/2024] [Accepted: 08/10/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND AND AIMS Severe acute respiratory syndrome coronavirus (SARS-CoV-2) preferentially infects the respiratory tract; however, several studies have implicated a multi-organ involvement. Hepatic dysfunctions caused by SARS-CoV-2 infection have been increasingly recognized and described to correlate with disease severity. To elucidate molecular factors that could contribute towards hepatic infection, we concentrated on microRNAs (miRNAs), a class of small non-coding RNAs that modulate various cellular processes and which are reported to be differentially regulated during liver injury. We aimed to study the infection of primary human hepatocytes (PHH) with SARS-CoV-2 and to evaluate the potential of miRNAs for modulating viral infection. METHODS We analysed liver autopsies from a coronavirus disease 19 (COVID-19)-positive cohort for the presence of viral RNA using Nanopore sequencing. PHH were used for the infection with SARS-CoV-2. The candidate miRNAs targeting angiotensin converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) were identified using in silico approaches. To discover the potential regulatory mechanism, transfection experiments, qRT-PCRs, western blots and luciferase reporter assays were performed. RESULTS We could detect SARS-CoV-2 RNA in COVID-19-positive liver autopsies. We show that PHH express ACE2 and TMPRSS2 and can be readily infected with SARS-CoV-2, resulting in robust replication. Transfection of selected miRNA mimics reduced SARS-CoV-2 receptor expression and SARS-CoV-2 burden in PHH. In silico and biochemical analyses supported a potential direct binding of miR-141-3p to the SARS-CoV-2 genome. CONCLUSION We confirm that PHH are susceptible to SARS-CoV-2 infection and demonstrate selected miRNAs targeting SARS-CoV-2 entry factors and/or the viral genome reduce viral loads. These data provide novel insights into hepatic susceptibility to SARS-CoV-2 and associated dysfunctions in COVID-19.
Collapse
Affiliation(s)
- Rajendra Khanal
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Research Group Liver Regeneration & RNA Therapeutics, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Natalie Heinen
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Alexandra Bogomolova
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Research Group Liver Regeneration & RNA Therapeutics, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Toni L Meister
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Institute for Infection Research and Vaccine Development (IIRVD), Centre for Internal Medicine, University Medical Centre Hamburg-Eppendorf (UKE), Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Simon T Herrmann
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Saskia Westhoven
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Maximilian K Nocke
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Daniel Todt
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- European Virus Bioinformatics Center (EVBC), Jena, Germany
| | - Freya Jockenhövel
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Isabel M Klein
- Tissue Bank of the German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Laura Hartmann
- Tissue Bank of the German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian W R Vondran
- Department of General, Visceral, Pediatric and Transplant Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Eike Steinmann
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Gert Zimmer
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Michael Ott
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Richard J P Brown
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Amar Deep Sharma
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Research Group Liver Regeneration & RNA Therapeutics, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Stephanie Pfaender
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
- University of Lübeck, Lübeck, Germany
| |
Collapse
|
22
|
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
|
23
|
Serwaa A, Oyawoye F, Owusu IA, Dosoo D, Manu AA, Sobo AK, Fosu K, Olwal CO, Quashie PK, Aikins AR. In vitro analysis suggests that SARS-CoV-2 infection differentially modulates cancer-like phenotypes and cytokine expression in colorectal and prostate cancer cells. Sci Rep 2024; 14:24625. [PMID: 39427065 PMCID: PMC11490510 DOI: 10.1038/s41598-024-75718-1] [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: 05/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) reportedly exacerbates cancer outcomes. However, how COVID-19 influences cancer prognosis and development remains poorly understood. Here, we investigated the effect of Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2), the etiological agent of COVID-19, on cellular cancer phenotypes the expression of cancer-related markers, and various proinflammatory cytokines. We infected prostate (22RV1) and colorectal (DLD-1) cancer cell lines, which express angiotensin-converting enzyme 2 (ACE2), with spike pseudovirus (sPV) and laboratory stocks of live SARS-CoV-2 viruses. After infection, we quantified changes in the cellular cancer phenotypes, the gene expression levels of some cancer markers, including Ki-67, BCL-2, VIM, MMP9, and VEGF, and proinflammatory cytokines. Phenotypic analysis was performed using MTT and wound healing assays, whereas gene expression analysis was carried out using real-time quantitative PCR (RT-qPCR). We show that SARS-CoV-2 infection impacts several key cellular pathways involved in cell growth, apoptosis, and migration, in prostate and colorectal cancer cells. Our results suggest that SARS-CoV-2 infection does influence various cancer cellular phenotypes and expression of molecular cancer markers and proinflammatory cytokines, albeit in a cell-type-specific manner. Our findings hint at the need for further studies and could have implications for evaluating the impact of other viruses on cancer progression.
Collapse
Affiliation(s)
- Alberta Serwaa
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Fatima Oyawoye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Irene Amoakoh Owusu
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Daniel Dosoo
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Aaron Adom Manu
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Augustine Kojo Sobo
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Kwadwo Fosu
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Charles Ochieng Olwal
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Peter Kojo Quashie
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.
- Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| | - Anastasia Rosebud Aikins
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.
| |
Collapse
|
24
|
Schermuly II, Romanet S, Patra AK, Mastrototaro L, Lemme A, Pieper R, Zentek J, Aschenbach JR. Transport of Neutral Amino Acids in the Jejunum of Pigs with Special Consideration of L-Methionine. Nutrients 2024; 16:3418. [PMID: 39408384 PMCID: PMC11478682 DOI: 10.3390/nu16193418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Methionine (Met) is a popular nutritional supplement in humans and animals. It is routinely supplemented to pigs as L-Met, DL-Met, or DL-2-hydroxy-4-(methylthio) butanoic acid (DL-HMTBA). Methods: We investigated the effect of these Met supplements on jejunal amino acid (AA) transport in male castrated Piétrain × Danbred pigs, also including a non-supplemented group. The mucosal-to-serosal flux of ten [14C]-labeled AAs (L-glutamine, glycine, L-leucine, L-lysine, L-Met, L-serine, L-threonine, L-tryptophan, L-tyrosine and L-valine) was investigated at two concentrations (50 µM and 5 mM). Inhibition of apical uptake by mucosal L-Met was also measured for these AAs. The intestinal expression of apical AA transporters, angiotensin-converting enzyme II and inflammation-related genes were compared with those of a previous study. Results: Except for tryptophan and lysine at 5 mM, all AA fluxes were Na+-dependent (p ≤ 0.05), and the uptake of most AAs, except glycine and lysine, was inhibited by L-Met (p < 0.001). A correlation network existed between Na+-dependent fluxes of most AAs (except tryptophan and partly glycine). We observed the upregulation of B0AT1 (SLC6A19) (p < 0.001), the downregulation of ATB0,+ (SLC6A14) (p < 0.001) and a lower expression of CASP1, IL1β, IL8, TGFβ and TNFα in the present vs. the previous study (p < 0.001). Conclusions: The correlating AAs likely share the same Na+-dependent transporter(s). A varying effect of the Met supplement type on AA transport in the two studies might be related to a different level of supplementation or a different inflammatory status of the small intestine.
Collapse
Affiliation(s)
- Isabel I. Schermuly
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| | - Stella Romanet
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| | - Amlan K. Patra
- American Institute for Goat Research, Langston University, Langston, OK 73050, USA;
| | - Lucia Mastrototaro
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - Andreas Lemme
- Animal Nutrition Services, Evonik Operations GmbH, Rodenbacher Chausee 4, 63457 Hanau-Wolfgang, Germany;
| | - Robert Pieper
- Institute of Animal Nutrition, Freie Universität Berlin, Königin-Luise-Straße 49, 14195 Berlin, Germany
| | - Jürgen Zentek
- Institute of Animal Nutrition, Freie Universität Berlin, Königin-Luise-Straße 49, 14195 Berlin, Germany
| | - Jörg R. Aschenbach
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| |
Collapse
|
25
|
Buibaş FI, Cercel RA, Şerbănescu MS, Turcu AA, Dumitrescu F, Pirici D, Marinescu I, Ionovici N, Busuioc CJ, Zorilă MV, Mogoantă L. Morphopathology of the lesions induced by SARS-CoV-2 infection in the lungs. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2024; 65:637-645. [PMID: 39957025 PMCID: PMC11924890 DOI: 10.47162/rjme.65.4.10] [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: 11/03/2024] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection spread rapidly from China around the world, causing the worst pandemic since the beginning of the 21st century. Although the disease named coronavirus disease 2019 (COVID-19) has multiple organ symptoms, the main pathological lesions occur in the lung, causing respiratory failure, pulmonary embolism, secondary bacterial pneumonia and pulmonary fibrosis. Despite the best efforts of researchers, the pathogenesis of SARS-CoV-2-induced cellular and tissue damage in organs and systems is poorly understood. Therefore, in our study, we aimed to highlight the pulmonary lesions and their extent, which could explain the complex symptomatology presented by patients who died with acute respiratory distress syndrome (ARDS). The study was performed on a number of 36 patients diagnosed with COVID-19 who died under legally suspicious conditions, requiring autopsy within the Romanian Forensic Medicine Institutes. All patients presented a local inflammatory reaction of pneumonic type, with exudative and proliferative phenomena, with intra-alveolar and interstitial inflammatory infiltrates formed by lymphocytes, macrophages and neutrophilic granulocytes, with congested or ruptured blood vessels with intra-alveolar or interstitial hemorrhages, with multiple thrombosis, with proliferation of local fibroblasts transformed into myofibroblasts and presence of granulation tissue that remodeled the entire lung parenchyma.
Collapse
|
26
|
Jia Y, Tian W, Li Y, Teng Y, Liu X, Li Z, Zhao M. Chloroquine: Rapidly withdrawing from first-line treatment of COVID-19. Heliyon 2024; 10:e37098. [PMID: 39281655 PMCID: PMC11402237 DOI: 10.1016/j.heliyon.2024.e37098] [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: 08/16/2023] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/18/2024] Open
Abstract
The COVID-19 outbreak has garnered significant global attention due to its impact on human health. Despite its relatively low fatality rate, the virus affects multiple organ systems, resulting in various symptoms such as palpitations, headaches, muscle pain, and hearing loss among COVID-19 patients and those recovering from the disease. These symptoms impose a substantial physical, psychological, and social burden on affected individuals. On February 15, 2020, the Chinese government advised incorporating antimalarial drugs into the guidelines issued by the National Health Commission of China for preventing, diagnosing, and treating COVID-19 pneumonia. We examine the adverse effects of Chloroquine (CQ) in treating COVID-19 complications to understand why it is no longer the primary treatment for the disease.
Collapse
Affiliation(s)
- Yunlong Jia
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Wenjie Tian
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Yuyao Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Yuyan Teng
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Xiaolin Liu
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Zhengyu Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Mingsheng Zhao
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| |
Collapse
|
27
|
V. J. A, P. J. A, T. M. A, Akhigbe RE. SARS-CoV-2 impairs male fertility by targeting semen quality and testosterone level: A systematic review and meta-analysis. PLoS One 2024; 19:e0307396. [PMID: 39250513 PMCID: PMC11383251 DOI: 10.1371/journal.pone.0307396] [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: 04/26/2024] [Accepted: 07/04/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Since the discovery of COVID-19 in December 2019, the novel virus has spread globally causing significant medical and socio-economic burden. Although the pandemic has been curtailed, the virus and its attendant complication live on. A major global concern is its adverse impact on male fertility. AIM This study was aimed to give an up to date and robust data regarding the effect of COVID-19 on semen variables and male reproductive hormones. MATERIALS AND METHODS Literature search was performed according to the recommendations of PRISMA. Out of the 852 studies collected, only 40 were eligible for inclusion in assessing the effect SARS-CoV-2 exerts on semen quality and androgens. More so, a SWOT analysis was conducted. RESULTS The present study demonstrated that SARS-CoV-2 significantly reduced ejaculate volume, sperm count, concentration, viability, normal morphology, and total and progressive motility. Furthermore, SARS-CoV-2 led to a reduction in circulating testosterone level, but a rise in oestrogen, prolactin, and luteinizing hormone levels. These findings were associated with a decline in testosterone/luteinizing hormone ratio. CONCLUSIONS The current study provides compelling evidence that SARS-CoV-2 may lower male fertility by reducing semen quality through a hormone-dependent mechanism; reduction in testosterone level and increase in oestrogen and prolactin levels.
Collapse
Affiliation(s)
- Ashonibare V. J.
- Medical Faculty, Department of Cardiovascular Surgery and Research Group for Experimental Surgery, Cardiovascular Regenerative Medicine and Tissue Engineering 3D Lab, Heinrich Heine University, Düsseldorf, Germany
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
| | - Ashonibare P. J.
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Akhigbe T. M.
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Agronomy, Breeding and Genetic Unit, Osun State University, Osun State, Nigeria
| | - R. E. Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| |
Collapse
|
28
|
Timofeeva AM, Nikitin AO, Nevinsky GA. Circulating miRNAs in the Plasma of Post-COVID-19 Patients with Typical Recovery and Those with Long-COVID Symptoms: Regulation of Immune Response-Associated Pathways. Noncoding RNA 2024; 10:48. [PMID: 39311385 PMCID: PMC11417918 DOI: 10.3390/ncrna10050048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024] Open
Abstract
Following the acute phase of SARS-CoV-2 infection, certain individuals experience persistent symptoms referred to as long COVID. This study analyzed the patients categorized into three distinct groups: (1) individuals presenting rheumatological symptoms associated with long COVID, (2) patients who have successfully recovered from COVID-19, and (3) donors who have never contracted COVID-19. A notable decline in the expression of miR-200c-3p, miR-766-3p, and miR-142-3p was identified among patients exhibiting rheumatological symptoms of long COVID. The highest concentration of miR-142-3p was found in healthy donors. One potential way to reduce miRNA concentrations is through antibody-mediated hydrolysis. Not only can antibodies possessing RNA-hydrolyzing activity recognize the miRNA substrate specifically, but they also catalyze its hydrolysis. The analysis of the catalytic activity of plasma antibodies revealed that antibodies from patients with long COVID demonstrated lower hydrolysis activity against five fluorescently labeled oligonucleotide sequences corresponding to the Flu-miR-146b-5p, Flu-miR-766-3p, Flu-miR-4742-3p, and Flu-miR-142-3p miRNAs and increased activity against the Flu-miR-378a-3p miRNA compared to other patient groups. The changes in miRNA concentrations and antibody-mediated hydrolysis of miRNAs are assumed to have a complex regulatory mechanism that is linked to gene pathways associated with the immune system. We demonstrate that all six miRNAs under analysis are associated with a large number of signaling pathways associated with immune response-associated pathways.
Collapse
Affiliation(s)
- Anna M. Timofeeva
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Artem O. Nikitin
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
| | - Georgy A. Nevinsky
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
29
|
Han SH, Ockerman K, Kirchmier M, Mardourian M, Bryan J, Cox E, Chim H, Spiguel L, Momeni A, Sorice-Virk S. Thrombotic Consequences of COVID-19 Infection on Microsurgical Reconstruction. Microsurgery 2024; 44:e31219. [PMID: 39207212 DOI: 10.1002/micr.31219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 06/12/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Evidence has shown increased morbidity and mortality for patients with COVID-19 infection within 7 weeks of surgery. However, no studies have specifically investigated the effects of COVID-19 in microsurgical outcomes. This study evaluated thrombotic and overall complications after free tissue transfer for a variety of indications in patients with and without previous COVID-19 infection. METHODS A retrospective cohort study was performed in adult patients with or without a history of COVID-19 infection who underwent microsurgical reconstruction between 2017 and 2022. Patients with a history of COVID-19 infection were matched to controls based on age, gender, race, body mass index, history of diabetes, coronary artery disease, hypertension, Caprini score, tobacco use, and flap indication. RESULTS From 2017 to 2022, 35 patients had a documented history of COVID-19. Matched case analysis determined a 4.8 times increased odds ratio of postoperative complications in the COVID-19 group compared with controls (p = 0.002). Significantly, more patients with COVID-19 experienced total or partial flap loss and anastomotic issues (COVID-19: 7/35, Control: 0/35; p < 0.001). There was no significant difference in incidence of VTE (COVID-19: 1/35, Control: 0/35; p = 0.493). Of note, 62.9% of the COVID-19 group were discharged on anticoagulants (versus 14.3% in the control group [p < 0.001]). CONCLUSION COVID-19 has dire, long-lasting effects on virtually every organ system, chief among them, the microcirculation. Further studies are needed to fully determine the extent and influence of COVID-19 on complex procedures such as free tissue transfer and how to optimize the screening, workup, and postoperative care to guard against the associated thrombotic consequences.
Collapse
Affiliation(s)
- Sabrina H Han
- Division of Plastic and Reconstructive Surgery, University of Chicago, Chicago, Illinois, USA
| | - Kyle Ockerman
- Division of Plastic and Reconstructive Surgery, University of Chicago, Chicago, Illinois, USA
| | - Matthew Kirchmier
- Division of Plastic and Reconstructive Surgery, University of Chicago, Chicago, Illinois, USA
| | - Markos Mardourian
- Division of Plastic and Reconstructive Surgery, University of Colorado, Aurora, Colorado, USA
| | - Jaimie Bryan
- Division of Plastic and Reconstructive Surgery, Stanford University, Palo Alto, California, USA
| | - Elizabeth Cox
- Division of Plastic and Reconstructive Surgery, Stanford University, Palo Alto, California, USA
| | - Harvey Chim
- Division of Plastic and Reconstructive Surgery, University of Florida, Gainesville, Florida, USA
| | - Lisa Spiguel
- Division of Surgical Oncology, University of Florida, Gainesville, Florida, USA
| | - Arash Momeni
- Division of Plastic and Reconstructive Surgery, Stanford University, Palo Alto, California, USA
| | - Sarah Sorice-Virk
- Division of Plastic and Reconstructive Surgery, Stanford University, Palo Alto, California, USA
| |
Collapse
|
30
|
Chatterjee S, Bhattacharya M, Saxena S, Lee SS, Chakraborty C. Autoantibodies in COVID-19 and Other Viral Diseases: Molecular, Cellular, and Clinical Perspectives. Rev Med Virol 2024; 34:e2583. [PMID: 39289528 DOI: 10.1002/rmv.2583] [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: 09/21/2023] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024]
Abstract
Autoantibodies are immune system-produced antibodies that wrongly target the body's cells and tissues for attack. The COVID-19 pandemic has made it possible to link autoantibodies to both the severity of pathogenic infection and the emergence of several autoimmune diseases after recovery from the infection. An overview of autoimmune disorders and the function of autoantibodies in COVID-19 and other infectious diseases are discussed in this review article. We also investigated the different categories of autoantibodies found in COVID-19 and other infectious diseases including the potential pathways by which they contribute to the severity of the illness. Additionally, it also highlights the probable connection between vaccine-induced autoantibodies and their adverse outcomes. The review also discusses the therapeutic perspectives of autoantibodies. This paper advances our knowledge about the intricate interaction between autoantibodies and COVID-19 by thoroughly assessing the most recent findings.
Collapse
Affiliation(s)
- Srijan Chatterjee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, South Korea
| | | | - Sanskriti Saxena
- Division of Biology, Indian Institute of Science Education and Research-Tirupati, Tirupati, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, South Korea
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| |
Collapse
|
31
|
Akpoviroro O, Sauers NK, Uwandu Q, Castagne M, Akpoviroro OP, Humayun S, Mirza W, Woodard J. Severe COVID-19 infection: An institutional review and literature overview. PLoS One 2024; 19:e0304960. [PMID: 39163410 PMCID: PMC11335168 DOI: 10.1371/journal.pone.0304960] [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: 07/17/2023] [Accepted: 05/21/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Our study aimed to describe the group of severe COVID-19 patients at an institutional level, and determine factors associated with different outcomes. METHODS A retrospective chart review of patients admitted with severe acute hypoxic respiratory failure due to COVID-19 infection. Based on outcomes, we categorized 3 groups of severe COVID-19: (1) Favorable outcome: progressive care unit admission and discharge (2) Intermediate outcome: ICU care (3) Poor outcome: in-hospital mortality. RESULTS Eighty-nine patients met our inclusion criteria; 42.7% were female. The average age was 59.7 (standard deviation (SD):13.7). Most of the population were Caucasian (95.5%) and non-Hispanic (91.0%). Age, sex, race, and ethnicity were similar between outcome groups. Medicare and Medicaid patients accounted for 62.9%. The average BMI was 33.5 (SD:8.2). Moderate comorbidity was observed, with an average Charlson Comorbidity index (CCI) of 3.8 (SD:2.6). There were no differences in the average CCI between groups(p = 0.291). Many patients (67.4%) had hypertension, diabetes (42.7%) and chronic lung disease (32.6%). A statistical difference was found when chronic lung disease was evaluated; p = 0.002. The prevalence of chronic lung disease was 19.6%, 27.8%, and 40% in the favorable, intermediate, and poor outcome groups, respectively. Smoking history was associated with poor outcomes (p = 0.04). Only 7.9% were fully vaccinated. Almost half (46.1%) were intubated and mechanically ventilated. Patients spent an average of 12.1 days ventilated (SD:8.5), with an average of 6.0 days from admission to ventilation (SD:5.1). The intermediate group had a shorter average interval from admission to ventilator (77.2 hours, SD:67.6), than the poor group (212.8 hours, SD:126.8); (p = 0.001). The presence of bacterial pneumonia was greatest in the intermediate group (72.2%), compared to the favorable group (17.4%), and the poor group (56%); this was significant (p<0.0001). In-hospital mortality was seen in 28.1%. CONCLUSION Most patients were male, obese, had moderate-level comorbidity, a history of tobacco abuse, and government-funded insurance. Nearly 50% required mechanical ventilation, and about 28% died during hospitalization. Bacterial pneumonia was most prevalent in intubated groups. Patients who were intubated with a good outcome were intubated earlier during their hospital course, with an average difference of 135.6 hours. A history of cigarette smoking and chronic lung disease were associated with poor outcomes.
Collapse
Affiliation(s)
- Ogheneyoma Akpoviroro
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, Pennsylvania, United States of America
| | - Nathan Kyle Sauers
- Department of Engineering, Pennsylvania State University, State College, Pennsylvania, United States of America
| | - Queeneth Uwandu
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, Pennsylvania, United States of America
| | - Myriam Castagne
- Clinical & Translational Science Institute, Boston University, Boston, Massachusetts, United States of America
| | | | - Sara Humayun
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, Pennsylvania, United States of America
| | - Wasique Mirza
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, Pennsylvania, United States of America
| | - Jameson Woodard
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, Pennsylvania, United States of America
| |
Collapse
|
32
|
Michaels TM, Essop MF, Joseph DE. Potential Effects of Hyperglycemia on SARS-CoV-2 Entry Mechanisms in Pancreatic Beta Cells. Viruses 2024; 16:1243. [PMID: 39205219 PMCID: PMC11358987 DOI: 10.3390/v16081243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The COVID-19 pandemic has revealed a bidirectional relationship between SARS-CoV-2 infection and diabetes mellitus. Existing evidence strongly suggests hyperglycemia as an independent risk factor for severe COVID-19, resulting in increased morbidity and mortality. Conversely, recent studies have reported new-onset diabetes following SARS-CoV-2 infection, hinting at a potential direct viral attack on pancreatic beta cells. In this review, we explore how hyperglycemia, a hallmark of diabetes, might influence SARS-CoV-2 entry and accessory proteins in pancreatic β-cells. We examine how the virus may enter and manipulate such cells, focusing on the role of the spike protein and its interaction with host receptors. Additionally, we analyze potential effects on endosomal processing and accessory proteins involved in viral infection. Our analysis suggests a complex interplay between hyperglycemia and SARS-CoV-2 in pancreatic β-cells. Understanding these mechanisms may help unlock urgent therapeutic strategies to mitigate the detrimental effects of COVID-19 in diabetic patients and unveil if the virus itself can trigger diabetes onset.
Collapse
Affiliation(s)
- Tara M. Michaels
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| | - M. Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa;
| | - Danzil E. Joseph
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| |
Collapse
|
33
|
Yesılyaprak T, Gok F. Examining the long-term effects of COVID-19 in surgical nurses: Case of Aegean Region. J Clin Nurs 2024; 33:3548-3555. [PMID: 38622927 DOI: 10.1111/jocn.17015] [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: 05/30/2022] [Revised: 12/10/2022] [Accepted: 01/07/2024] [Indexed: 04/17/2024]
Abstract
PURPOSE To examine the long-term effects of COVID-19 on surgical nurses. BACKGROUND Individuals contaminated with COVID-19 may face several metabolic or psychological issues, primarily in the respiratory, cardiovascular, nervous, musculoskeletal and renal systems during the late period. However, the long-term epidemiology is still not clear. DESIGN Descriptive cross-sectional study. METHODS The study included nurses (n = 509) who had been diagnosed with COVID-19 at least 12 weeks before and worked in surgical departments. We collected the study data via an online survey using the snowball sampling method between December 2021 and May 2022. This study followed the Reporting of Observational Studies in Epidemiology Guideline. RESULTS The mean age of the nurses was 31.66 ± 8.74 years. Nurses stated that they were diagnosed with COVID-19 approximately 36 weeks before participating in this study. We found that the nurses mostly experienced palpitation (83.5%), headache (73.5%), dyspnea (64.1%), anosmia (57.6%), arthralgia (55.7%) and burnout (58.4%) during the late period after COVID-19. CONCLUSION The long-term effects of COVID-19 were related to multiple organ dysfunctions. NO PATIENT OR PUBLIC CONTRIBUTION Since the study was conducted with healthy individuals who had previously experienced COVID-19, there is no patient contribution. RELEVANCE TO CLINICAL PRACTICE This study focuses on the long-term effects of COVID-19 on nurses. The results support the long-term effects of COVID-19 and are thought to contribute to the literature.
Collapse
Affiliation(s)
- Tugce Yesılyaprak
- Denizli Vocational School of Health Services, Pamukkale University, Pamukkale, Turkey
| | - Fadime Gok
- Faculty of Health Sciences, Pamukkale University, Pamukkale, Turkey
| |
Collapse
|
34
|
Mohamed RH, Abdel Hay NH, Fawzy NM, Tamim YM, Doaa Karem MM, Yehia DAY, Abdel Maksoud OM, Abdelrahim DS. Targeting mevalonate pathway by zoledronate ameliorated pulmonary fibrosis in a rat model: Promising therapy against post-COVID-19 pulmonary fibrosis. Fundam Clin Pharmacol 2024; 38:703-717. [PMID: 38357833 DOI: 10.1111/fcp.12994] [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: 09/23/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Rho kinase (ROCK) pathway plays a critical role in post-COVID-19 pulmonary fibrosis (PCPF) and its intervention with angiotensin-converting enzyme 2 (ACE2) and vascular endothelial growth factor (VEGF) will be a potential therapeutic target. OBJECTIVES The present study was conducted to investigate the efficacy of zoledronate (ZA) on carbon tetrachloride (CCl4) induced pulmonary fibrosis (PF) in rats through targeting ACE2, ROCK, and VEGF signaling pathways. METHODS Fifty male Wistar rats were divided into five groups: control, vehicle-treated, PF, PF-ZA 50, and PF-ZA 100 groups. ZA was given in two different doses 100 and 50 μg/kg/week intraperitoneally. After anesthesia, mean arterial blood pressure (MBP) was measured. After scarification, lung coefficient was calculated. Lung levels of ACE 2, interleukin-1β (IL-1β), transforming growth factor-β (TGF-β), VEGF, glutathione (GSH), and superoxide dismutase (SOD) were measured. Expression of ROCK, phosphorylated myosin phosphatase target subunit 1 (P-MYPT1), and matrix metalloproteinase (MMP-1), along with histopathological changes and immune-histochemical staining for lung α-smooth muscle actin (α-SMA), tumor necrosis factor-alpha (TNFα), and caspase-3, were evaluated. RESULTS ZA significantly prevented the decrease in MBP. ZA significantly increased ACE2, GSH, and SOD and significantly decreased IL-1β, TGF-β, and VEGF in lung in comparison to PF group. ZA prevented the histopathological changes induced by CCl4. ZA inhibited lung expression of ROCK, P-MYPT1, MMP-1, α-SMA, TNFα, and caspase-3 with significant differences favoring the high dose intervention. CONCLUSION ZA in a dose-dependent manner prevented the pathological effect of CCl4 in the lung by targeting mevalonate pathway. It could be promising therapy against PCPF.
Collapse
Affiliation(s)
- Reham Hussein Mohamed
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nesma Hussein Abdel Hay
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nesma Mohamed Fawzy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Yomna M Tamim
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - M M Doaa Karem
- Department of Histology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Omnia M Abdel Maksoud
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Dina S Abdelrahim
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Department of Pharmacology, Faculty of Medicine, Modern Technology and Information University, Cairo, Egypt
| |
Collapse
|
35
|
AbuSaleh L, Ereqat S, Al-Jawabreh A, Nasereddin A. Genetic Polymorphisms of Angiotensin-Converting Enzyme 1 (ACE1) and ACE2 Associated With Severe Acute Respiratory Syndrome COVID-19 in the Palestinian Population. Cureus 2024; 16:e67670. [PMID: 39318909 PMCID: PMC11420599 DOI: 10.7759/cureus.67670] [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] [Accepted: 08/23/2024] [Indexed: 09/26/2024] Open
Abstract
As a key enzyme of the renin-angiotensin system (RAS), angiotensin-converting enzyme 2 (ACE2) is a validated receptor for SARS-CoV-2, linking RAS to COVID-19. Functional ACE1/ACE2 gene polymorphisms likely cause an imbalance in the ACE1/ACE2 ratio, triggering RAS imbalance and may contribute to COVID-19 complications. This study aimed to investigate four single nucleotide polymorphisms (SNPs) of ACE1 and ACE2 genes, three for ACE1 (rs4343, rs4342, rs4341) and one for ACE2 (rs2285666), in patients with COVID-19 among the Palestinian population. A total of 130 blood samples were collected, including 50 negative controls without COVID-19 infection, 50 cases with COVID-19 infection but not hospitalized, and 30 patients with severe COVID-19 infection hospitalized in the intensive care unit. Fragments of the ACE1 and ACE2 genes, including the targeted SNPs, were amplified using multiplex PCR and subsequently genotyped by next-generation sequencing with specific virtual probes. Our results revealed that ACE2 rs2285666 GG genotype carriers were more prevalent in COVID-19 patients compared to the control group (P=0.049), while no statistical differences were observed in the distribution of ACE1 (rs4343, rs4342, rs4341) variants between COVID-19 patients and the control group. GA carriers of ACE2, rs2285666, among cases and ICU groups were at lower risk of getting COVID-19 infection (P=0.002 and P=0.013, respectively), and they were unlikely to develop fatigue (P=0.043), headache (P=0.007), loss of smell (P=0.028), and dyspnea (P=0.005). Age and comorbidities such as hypertension and coronary artery disease (CAD) were independent risk factors for COVID-19 disease. Symptoms of COVID-19 patients such as fatigue, headaches, runny noses, and loss of smell were significantly higher in non-hospitalized cases of COVID-19, while dyspnea was more frequent in the ICU patients. In conclusion, these findings indicate that the ACE2 rs2285666 GG genotype is associated with an increased risk of COVID-19 infection. This association suggests a potential genetic predisposition linked to the ACE2 gene, which may influence the susceptibility and severity of the disease.
Collapse
Affiliation(s)
- Lama AbuSaleh
- Biochemistry and Molecular Biology Department, Faculty of Medicine, Al-Quds University, Jerusalem, PSE
| | - Suheir Ereqat
- Biochemistry and Molecular Biology Department, Al-Quds University, Jerusalem, PSE
| | - Amer Al-Jawabreh
- Medical Laboratory Sciences Department, Faculty of Allied Health Sciences, Arab American University, Jerusalem, PSE
| | | |
Collapse
|
36
|
Espinoza-Gutiérrez HA, López-Salido SC, Flores-Soto ME, Tejeda-Martínez AR, Chaparro-Huerta V, Viveros-Paredes JM. Angiotensinergic effect of β-Caryophyllene on Lipopolysaccharide- induced systemic inflammation. Biochem Biophys Res Commun 2024; 719:150081. [PMID: 38744071 DOI: 10.1016/j.bbrc.2024.150081] [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: 03/07/2024] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
Renin-Angiotensin System (RAS) is a peptidergic system, canonically known for its role in blood pressure regulation. Furthermore, a non-canonical RAS regulates pathophysiological phenomena, such as inflammation since it consists of two main axes: the pro-inflammatory renin/(pro)renin receptor ((P)RR) axis, and the anti-inflammatory angiotensin-converting enzyme 2 (ACE2)/Angiotensin-(1-7) (Ang-(1-7))/Mas Receptor (MasR) axis. Few phytochemicals have shown to exert angiotensinergic and anti-inflammatory effects through some of these axes; nevertheless, anti-inflammatory drugs, such as phytocannabinoids have not been studied regarding this subject. Among phytocannabinoids, β-Caryophyllene stands out as a dietary phytocannabinoid with antiphlogistic activity that possess a unique sesquiterpenoid structure. Although its cannabinergic effect has been studied, its angiotensinergic effect reminds underexplored. This study aims to explore the angiotensinergic effect of β-Caryophyllene on inflammation and stress at a systemic level. After intranasal Lipopolysaccharide (LPS) installation and oral treatment with β-Caryophyllene, the concentration and activity of key RAS elements in the serum, such as Renin, ACE2 and Ang-(1-7), along with the stress hormone corticosterone and pro/anti-inflammatory cytokines, were measured in mice serum. The results show that β-Caryophyllene treatment modified RAS levels by increasing Renin and Ang-(1-7), alongside the reduction of pro-inflammatory cytokines and corticosterone levels. These results indicate that β-Caryophyllene exhibits angiotensinergic activity in favor of anti-inflammation.
Collapse
Affiliation(s)
- Hugo Alejandro Espinoza-Gutiérrez
- Laboratorio de Investigación y Desarrollo Farmacéutico, Departamento de Farmacología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, 44430, Guadalajara, Jalisco, Mexico; Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano Del Seguro Social, 44340, Guadalajara, Jalisco, Mexico
| | - Sofía Cecilia López-Salido
- Laboratorio de Investigación y Desarrollo Farmacéutico, Departamento de Farmacología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, 44430, Guadalajara, Jalisco, Mexico; Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano Del Seguro Social, 44340, Guadalajara, Jalisco, Mexico
| | - Mario Eduardo Flores-Soto
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano Del Seguro Social, 44340, Guadalajara, Jalisco, Mexico
| | - Aldo Rafael Tejeda-Martínez
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano Del Seguro Social, 44340, Guadalajara, Jalisco, Mexico
| | - Veronica Chaparro-Huerta
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano Del Seguro Social, 44340, Guadalajara, Jalisco, Mexico
| | - Juan Manuel Viveros-Paredes
- Laboratorio de Investigación y Desarrollo Farmacéutico, Departamento de Farmacología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, 44430, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
37
|
Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function. Int J Mol Sci 2024; 25:7553. [PMID: 39062796 PMCID: PMC11277036 DOI: 10.3390/ijms25147553] [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: 05/27/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Proteases are produced and released in the mucosal cells of the respiratory tract and have important physiological functions, for example, maintaining airway humidification to allow proper gas exchange. The infectious mechanism of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), takes advantage of host proteases in two ways: to change the spatial conformation of the spike (S) protein via endoproteolysis (e.g., transmembrane serine protease type 2 (TMPRSS2)) and as a target to anchor to epithelial cells (e.g., angiotensin-converting enzyme 2 (ACE2)). This infectious process leads to an imbalance in the mucosa between the release and action of proteases versus regulation by anti-proteases, which contributes to the exacerbation of the inflammatory and prothrombotic response in COVID-19. In this article, we describe the most important proteases that are affected in COVID-19, and how their overactivation affects the three main physiological systems in which they participate: the complement system and the kinin-kallikrein system (KKS), which both form part of the contact system of innate immunity, and the renin-angiotensin-aldosterone system (RAAS). We aim to elucidate the pathophysiological bases of COVID-19 in the context of the imbalance between the action of proteases and anti-proteases to understand the mechanism of aprotinin action (a panprotease inhibitor). In a second-part review, titled "Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions", we explain in depth the pharmacodynamics, pharmacokinetics, toxicity, and use of aprotinin as an antiviral drug.
Collapse
Affiliation(s)
- Juan Fernando Padín
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
| | - José Manuel Pérez-Ortiz
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco Javier Redondo-Calvo
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
- Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
- Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
| |
Collapse
|
38
|
Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions. Int J Mol Sci 2024; 25:7209. [PMID: 39000315 PMCID: PMC11241800 DOI: 10.3390/ijms25137209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Aprotinin is a broad-spectrum inhibitor of human proteases that has been approved for the treatment of bleeding in single coronary artery bypass surgery because of its potent antifibrinolytic actions. Following the outbreak of the COVID-19 pandemic, there was an urgent need to find new antiviral drugs. Aprotinin is a good candidate for therapeutic repositioning as a broad-spectrum antiviral drug and for treating the symptomatic processes that characterise viral respiratory diseases, including COVID-19. This is due to its strong pharmacological ability to inhibit a plethora of host proteases used by respiratory viruses in their infective mechanisms. The proteases allow the cleavage and conformational change of proteins that make up their viral capsid, and thus enable them to anchor themselves by recognition of their target in the epithelial cell. In addition, the activation of these proteases initiates the inflammatory process that triggers the infection. The attraction of the drug is not only its pharmacodynamic characteristics but also the possibility of administration by the inhalation route, avoiding unwanted systemic effects. This, together with the low cost of treatment (≈2 Euro/dose), makes it a good candidate to reach countries with lower economic means. In this article, we will discuss the pharmacodynamic, pharmacokinetic, and toxicological characteristics of aprotinin administered by the inhalation route; analyse the main advances in our knowledge of this medication; and the future directions that should be taken in research in order to reposition this medication in therapeutics.
Collapse
Affiliation(s)
- Juan-Fernando Padín
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain
| | - José Manuel Pérez-Ortiz
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco Javier Redondo-Calvo
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain
- Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
- Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
| |
Collapse
|
39
|
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
|
40
|
Li X, Mi Z, Liu Z, Rong P. SARS-CoV-2: pathogenesis, therapeutics, variants, and vaccines. Front Microbiol 2024; 15:1334152. [PMID: 38939189 PMCID: PMC11208693 DOI: 10.3389/fmicb.2024.1334152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/29/2024] [Indexed: 06/29/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in December 2019 with staggering economic fallout and human suffering. The unique structure of SARS-CoV-2 and its underlying pathogenic mechanism were responsible for the global pandemic. In addition to the direct damage caused by the virus, SARS-CoV-2 triggers an abnormal immune response leading to a cytokine storm, culminating in acute respiratory distress syndrome and other fatal diseases that pose a significant challenge to clinicians. Therefore, potential treatments should focus not only on eliminating the virus but also on alleviating or controlling acute immune/inflammatory responses. Current management strategies for COVID-19 include preventative measures and supportive care, while the role of the host immune/inflammatory response in disease progression has largely been overlooked. Understanding the interaction between SARS-CoV-2 and its receptors, as well as the underlying pathogenesis, has proven to be helpful for disease prevention, early recognition of disease progression, vaccine development, and interventions aimed at reducing immunopathology have been shown to reduce adverse clinical outcomes and improve prognosis. Moreover, several key mutations in the SARS-CoV-2 genome sequence result in an enhanced binding affinity to the host cell receptor, or produce immune escape, leading to either increased virus transmissibility or virulence of variants that carry these mutations. This review characterizes the structural features of SARS-CoV-2, its variants, and their interaction with the immune system, emphasizing the role of dysfunctional immune responses and cytokine storm in disease progression. Additionally, potential therapeutic options are reviewed, providing critical insights into disease management, exploring effective approaches to deal with the public health crises caused by SARS-CoV-2.
Collapse
Affiliation(s)
- Xi Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ze Mi
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhenguo Liu
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
41
|
Carvajal JJ, García-Castillo V, Cuellar SV, Campillay-Véliz CP, Salazar-Ardiles C, Avellaneda AM, Muñoz CA, Retamal-Díaz A, Bueno SM, González PA, Kalergis AM, Lay MK. New insights into the pathogenesis of SARS-CoV-2 during and after the COVID-19 pandemic. Front Immunol 2024; 15:1363572. [PMID: 38911850 PMCID: PMC11190347 DOI: 10.3389/fimmu.2024.1363572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 04/24/2024] [Indexed: 06/25/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the respiratory distress condition known as COVID-19. This disease broadly affects several physiological systems, including the gastrointestinal, renal, and central nervous (CNS) systems, significantly influencing the patient's overall quality of life. Additionally, numerous risk factors have been suggested, including gender, body weight, age, metabolic status, renal health, preexisting cardiomyopathies, and inflammatory conditions. Despite advances in understanding the genome and pathophysiological ramifications of COVID-19, its precise origins remain elusive. SARS-CoV-2 interacts with a receptor-binding domain within angiotensin-converting enzyme 2 (ACE2). This receptor is expressed in various organs of different species, including humans, with different abundance. Although COVID-19 has multiorgan manifestations, the main pathologies occur in the lung, including pulmonary fibrosis, respiratory failure, pulmonary embolism, and secondary bacterial pneumonia. In the post-COVID-19 period, different sequelae may occur, which may have various causes, including the direct action of the virus, alteration of the immune response, and metabolic alterations during infection, among others. Recognizing the serious adverse health effects associated with COVID-19, it becomes imperative to comprehensively elucidate and discuss the existing evidence surrounding this viral infection, including those related to the pathophysiological effects of the disease and the subsequent consequences. This review aims to contribute to a comprehensive understanding of the impact of COVID-19 and its long-term effects on human health.
Collapse
Affiliation(s)
- Jonatan J. Carvajal
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Valeria García-Castillo
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Shelsy V. Cuellar
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | | | - Camila Salazar-Ardiles
- Center for Research in Physiology and Altitude Medicine (FIMEDALT), Biomedical Department, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Andrea M. Avellaneda
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Department of Basic Sciences, Faculty of Sciences, Universidad Santo Tomás, Antofagasta, Chile
| | - Christian A. Muñoz
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Angello Retamal-Díaz
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K. Lay
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| |
Collapse
|
42
|
Tsukamoto A, Jae Man L, Oyama K, Masuda A, Mon H, Ueda T, Kusakabe T. Effective expression and characterization of the receptor binding domains in SARS-CoV-2 Spike proteins from original strain and variants of concern using Bombyx mori nucleopolyhedrovirus in silkworm. Protein Expr Purif 2024; 218:106450. [PMID: 38395208 DOI: 10.1016/j.pep.2024.106450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
A new coronavirus, known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is responsible for the global pandemic of COVID-19 in 2020. Through structural analysis, it was found that several amino acid residues in the human angiotensin-converting enzyme-2 (hACE2) receptor directly interact with those in the receptor binding domain (RBD) of the spike glycoprotein (S-protein). Various cell lines, including HEK293, HeLa cells, and the baculovirus expression vector system (BEVS) with the insect cell line Sf9, have been utilized to produce the RBD. In this study, we investigated the use of Bombyx mori nucleopolyhedrovirus (BmNPV) and BEVS. For efficient production of a highly pure recombinant RBD protein, we designed it with two tags (His tag and STREP tag) at the C-terminus and a solubilizing tag (SUMO) at the N-terminus. After expressing the protein using BmNPV and silkworm and purifying it with a HisTrap excel column, the eluted protein was digested with SUMO protease and further purified using a Strep-Tactin Superflow column. As a result, we obtained the RBD as a monomer with a yield of 2.6 mg/10 mL serum (equivalent to 30 silkworms). The RBD showed an affinity for the hACE2 receptor. Additionally, the RBDs from the Alpha, Beta, Gamma, Delta, and Omicron variants were expressed and purified using the same protocol. It was found that the RBD from the Alpha, Beta, Gamma, and Delta variants could be obtained with yields of 1.4-2.6 mg/10 mL serum and had an affinity to the hACE2 receptor.
Collapse
Affiliation(s)
- Akira Tsukamoto
- Graduate School of Pharmaceutical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Lee Jae Man
- Laboratory of Creative Science for Insect Industries, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Kosuke Oyama
- Graduate School of Pharmaceutical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akitsu Masuda
- Laboratory of Creative Science for Insect Industries, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Hiroaki Mon
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Tadashi Ueda
- Graduate School of Pharmaceutical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Takahiro Kusakabe
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
| |
Collapse
|
43
|
Al‐Qahtani Z, Al‐kuraishy HM, Al‐Gareeb AI, Albuhadily AK, Ali NH, Alexiou A, Papadakis M, Saad HM, Batiha GE. The potential role of brain renin-angiotensin system in the neuropathology of Parkinson disease: Friend, foe or turncoat? J Cell Mol Med 2024; 28:e18495. [PMID: 38899551 PMCID: PMC11187740 DOI: 10.1111/jcmm.18495] [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: 11/04/2023] [Revised: 01/15/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Parkinson disease (PD) is one of the most common neurodegenerative diseases of the brain. Of note, brain renin-angiotensin system (RAS) is intricate in the PD neuropathology through modulation of oxidative stress, mitochondrial dysfunction and neuroinflammation. Therefore, modulation of brain RAS by angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEIs) may be effective in reducing the risk and PD neuropathology. It has been shown that all components including the peptides and enzymes of the RAS are present in the different brain areas. Brain RAS plays a critical role in the regulation of memory and cognitive function, and in the controlling of central blood pressure. However, exaggerated brain RAS is implicated in the pathogenesis of different neurodegenerative diseases including PD. Two well-known pathways of brain RAS are recognized including; the classical pathway which is mainly mediated by AngII/AT1R has detrimental effects. Conversely, the non-classical pathway which is mostly mediated by ACE2/Ang1-7/MASR and AngII/AT2R has beneficial effects against PD neuropathology. Exaggerated brain RAS affects the viability of dopaminergic neurons. However, the fundamental mechanism of brain RAS in PD neuropathology was not fully elucidated. Consequently, the purpose of this review is to disclose the mechanistic role of RAS in in the pathogenesis of PD. In addition, we try to revise how the ACEIs and ARBs can be developed for therapeutics in PD.
Collapse
Affiliation(s)
- Zainah Al‐Qahtani
- Neurology Section, Internal Medicine Department, College of MedicineKing khaled universityAbhaSaudi Arabia
| | - Hayder M. Al‐kuraishy
- Clinical pharmacology and medicine, college of medicineMustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Clinical pharmacology and medicine, college of medicineMustansiriyah UniversityBaghdadIraq
| | - Ali K. Albuhadily
- Clinical pharmacology and medicine, college of medicineMustansiriyah UniversityBaghdadIraq
| | - Naif H. Ali
- Department of Internal Medicine, Medical CollegeNajran UniversityNajranSaudi Arabia
| | - Athanasios Alexiou
- University Centre for Research & DevelopmentChandigarh UniversityMohaliIndia
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- Department of Research & Development, FunogenAthensGreece
- Department of Research & DevelopmentAFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐HerdeckeWuppertalGermany
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMatrouhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourAlBeheiraEgypt
| |
Collapse
|
44
|
Hu W, Tan J, Lin Y, Tao Y, Zhou Q. Bibliometric and visual analysis of ACE2/Ang 1-7/MasR axis in diabetes and its microvascular complications from 2000 to 2023. Heliyon 2024; 10:e31405. [PMID: 38807880 PMCID: PMC11130665 DOI: 10.1016/j.heliyon.2024.e31405] [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: 01/19/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/30/2024] Open
Abstract
Background The pathogenesis of diabetes and its microvascular complications are intimately associated with renin angiotensin system dysregulation. Evidence suggests the angiotensin converting enzyme 2 (ACE2)/angiotensin 1-7 (Ang 1-7)/Mas receptor (MasR) axis regulates metabolic imbalances, inflammatory responses, reduces oxidative stress, and sustains microvascular integrity, thereby strengthening defences against diabetic conditions. This study aims to conduct a comprehensive analysis of the ACE2/Ang 1-7/MasR axis in diabetes and its microvascular complications over the past two decades, focusing on key contributors, research hotspots, and thematic trends. Methods This cross-sectional bibliometric analysis of 349 English-language publications was performed using HistCite, VOSviewer, CiteSpace, and Bibliometrix R for visualization and metric analysis. Primary analytical metrics included publication count and keyword trend dynamics. Results The United States, contributing 105 articles, emerged as the most productive country, with the University of Florida leading institutions with 18 publications. Benter IF was the most prolific author with 14 publications, and Clinical Science was the leading journal with 13 articles. A total of 151 of the 527 author's keywords with two or more occurrences clustered into four major clusters: diabetic microvascular pathogenesis, metabolic systems, type 2 diabetes, and coronavirus infections. Keywords such as "SARS", "ACE2", "coronavirus", "receptor" and "infection" displayed the strongest citation bursts. The thematic evolution in this field expanded from focusing on the renin angiotensin system (2002-2009) to incorporating ACE2 and diabetes metabolism (2010-2016). The latter period (2017-2023) witnessed a significant surge in diabetes research, reflecting the impact of COVID-19 and associated conditions such as diabetic retinopathy and cardiomyopathy. Conclusions This scientometric study offers a detailed analysis of the ACE2/Ang 1-7/MasR axis in diabetes and its microvascular complications, providing valuable insights for future research directions.
Collapse
Affiliation(s)
- Weiwen Hu
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Jian Tan
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Yeting Lin
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Yulin Tao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Qiong Zhou
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| |
Collapse
|
45
|
Luo D, Bai M, Zhang W, Wang J. The possible mechanism and research progress of ACE2 involved in cardiovascular injury caused by COVID-19: a review. Front Cardiovasc Med 2024; 11:1409723. [PMID: 38863899 PMCID: PMC11165996 DOI: 10.3389/fcvm.2024.1409723] [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: 03/30/2024] [Accepted: 05/09/2024] [Indexed: 06/13/2024] Open
Abstract
ACE2 is the earliest receptor discovered to mediate the entry of SARS-CoV-2. In addition to the receptor, it also participates in complex pathological and physiological processes, including regulating the RAS system, apelin, KKS system, and immune system. In addition to affecting the respiratory system, viral infections also interact with cardiovascular diseases. SARS-CoV-2 can directly invade the cardiovascular system through ACE2; Similarly, cardiovascular diseases such as hypertension and coronary heart disease can affect ACE2 levels and exacerbate the disease, and ACE2 dysregulation may also be a potential mechanism for long-term acute sequelae of COVID-19. Since the SARS CoV-2 epidemic, many large population studies have tried to clarify the current focus of debate, that is, whether we should give COVID-19 patients ACEI and ARB drug treatment, but there is still no conclusive conclusion. We also discussed potential disease treatment options for ACE2 at present. Finally, we discussed the researchers' latest findings on ACE2 and their prospects for future research.
Collapse
Affiliation(s)
| | | | | | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
46
|
Yang C, Tan Y, Li Z, Hu L, Chen Y, Zhu S, Hu J, Huai T, Li M, Zhang G, Rao D, Fei G, Shao M, Ding Z. Pulmonary redox imbalance drives early fibroproliferative response in moderate/severe coronavirus disease-19 acute respiratory distress syndrome and impacts long-term lung abnormalities. Ann Intensive Care 2024; 14:72. [PMID: 38735020 PMCID: PMC11089033 DOI: 10.1186/s13613-024-01293-3] [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: 12/09/2023] [Accepted: 04/10/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND COVID-19-associated pulmonary fibrosis remains frequent. This study aimed to investigate pulmonary redox balance in COVID-19 ARDS patients and possible relationship with pulmonary fibrosis and long-term lung abnormalities. METHODS Baseline data, chest CT fibrosis scores, N-terminal peptide of alveolar collagen III (NT-PCP-III), transforming growth factor (TGF)-β1, superoxide dismutase (SOD), reduced glutathione (GSH), oxidized glutathione (GSSG) and malondialdehyde (MDA) in bronchoalveolar lavage fluid (BALF) were first collected and compared between SARS-CoV-2 RNA positive patients with moderate to severe ARDS (n = 65, COVID-19 ARDS) and SARS-CoV-2 RNA negative non-ARDS patients requiring mechanical ventilation (n = 63, non-ARDS). Then, correlations between fibroproliferative (NT-PCP-III and TGF-β1) and redox markers were analyzed within COVID-19 ARDS group, and comparisons between survivor and non-survivor subgroups were performed. Finally, follow-up of COVID-19 ARDS survivors was performed to analyze the relationship between pulmonary abnormalities, fibroproliferative and redox markers 3 months after discharge. RESULTS Compared with non-ARDS group, COVID-19 ARDS group had significantly elevated chest CT fibrosis scores (p < 0.001) and NT-PCP-III (p < 0.001), TGF-β1 (p < 0.001), GSSG (p < 0.001), and MDA (p < 0.001) concentrations on admission, while decreased SOD (p < 0.001) and GSH (p < 0.001) levels were observed in BALF. Both NT-PCP-III and TGF-β1 in BALF from COVID-19 ARDS group were directly correlated with GSSG (p < 0.001) and MDA (p < 0.001) and were inversely correlated with SOD (p < 0.001) and GSH (p < 0.001). Within COVID-19 ARDS group, non-survivors (n = 28) showed significant pulmonary fibroproliferation (p < 0.001) with more severe redox imbalance (p < 0.001) than survivors (n = 37). Furthermore, according to data from COVID-19 ARDS survivor follow-up (n = 37), radiographic residual pulmonary fibrosis and lung function impairment improved 3 months after discharge compared with discharge (p < 0.001) and were associated with early pulmonary fibroproliferation and redox imbalance (p < 0.01). CONCLUSIONS Pulmonary redox imbalance occurring early in COVID-19 ARDS patients drives fibroproliferative response and increases the risk of death. Long-term lung abnormalities post-COVID-19 are associated with early pulmonary fibroproliferation and redox imbalance.
Collapse
Affiliation(s)
- Chun Yang
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Yuanyuan Tan
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Zihao Li
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Lei Hu
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Yuanyuan Chen
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Shouliang Zhu
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Jiawei Hu
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Tingting Huai
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Mingqing Li
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Guobin Zhang
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Dewang Rao
- Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Guanghe Fei
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China.
| | - Min Shao
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China.
| | - Zhenxing Ding
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China.
| |
Collapse
|
47
|
Amin R, Sohrabi MR, Zali AR, Hannani K. COVID-19 in pregnancy: A cross-sectional study on clinical features, disease severity, and health outcome. BIOMOLECULES & BIOMEDICINE 2024; 24:659-664. [PMID: 38315087 PMCID: PMC11088891 DOI: 10.17305/bb.2023.9748] [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: 09/01/2023] [Revised: 10/07/2023] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
Assessing the impact of coronavirus disease 2019 (COVID-19) reveals unique challenges for pregnant women, who experience distinct clinical manifestations and health outcomes compared to their non-pregnant counterparts. We aimed to evaluate the clinical features, disease severity, and health outcomes of COVID-19 in pregnant women and compare them to those of non pregnant women. In this population-based study, we included all women diagnosed with COVID-19 across the province of Tehran during the first two years of the epidemic. Descriptive statistics, the chi-squared test, and the logistic regression model were applied. Overall, 79,338 non-pregnant women and 3249 pregnant women diagnosed with COVID-19 were included. Pregnant women were most commonly in the age group of 25 - 34 years (54%, n = 1758), while the age group of 34-44 had the highest representation among non-pregnant women (56%, n = 44,492). After accounting for age and comorbidities, pregnancy was associated with an increased risk of requiring intensive care (odds ratio [OR] 1.38, confidence interval [CI] 1.223 - 1.564). However, the probability of dying due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection was lower in pregnant women compared to non-pregnant women (OR 0.55, CI 0.394-0.793). Cough (41%) and fever (30%) were the most frequent clinical presentations in pregnant women, whereas cough (57%) and muscle ache (38%) were the most common symptoms in non-pregnant women. Furthermore, diarrhea (P < 0.001) and skin lesions (P < 0.001) were reported more frequently by pregnant patients than non-pregnant patients. A significant prevalence of diabetes (P < 0.001), hypertension (P < 0.001), cancers (P < 0.001), and chronic hematological diseases (P < 0.001) was observed in pregnant patients. In conclusion, COVID-19-infected pregnant women exhibit different clinical manifestations and a more severe clinical course but have better health outcomes compared to their non-pregnant counterparts.
Collapse
Affiliation(s)
- Rozhin Amin
- Community Medicine Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Social Determinants of Health Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Sohrabi
- Community Medicine Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Social Determinants of Health Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali-Reza Zali
- Functional Neurosurgery Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khatereh Hannani
- Statistics and Information Technology Management, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
48
|
Ceasovschih A, Șorodoc V, Covantsev S, Balta A, Uzokov J, Kaiser SE, Almaghraby A, Lionte C, Stătescu C, Sascău RA, Onofrei V, Haliga RE, Stoica A, Bologa C, Ailoaei Ș, Şener YZ, Kounis NG, Șorodoc L. Electrocardiogram Features in Non-Cardiac Diseases: From Mechanisms to Practical Aspects. J Multidiscip Healthc 2024; 17:1695-1719. [PMID: 38659633 PMCID: PMC11041971 DOI: 10.2147/jmdh.s445549] [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: 12/18/2023] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
Despite the noteworthy advancements and the introduction of new technologies in diagnostic tools for cardiovascular disorders, the electrocardiogram (ECG) remains a reliable, easily accessible, and affordable tool to use. In addition to its crucial role in cardiac emergencies, ECG can be considered a very useful ancillary tool for the diagnosis of many non-cardiac diseases as well. In this narrative review, we aimed to explore the potential contributions of ECG for the diagnosis of non-cardiac diseases such as stroke, migraine, pancreatitis, Kounis syndrome, hypothermia, esophageal disorders, pulmonary embolism, pulmonary diseases, electrolyte disturbances, anemia, coronavirus disease 2019, different intoxications and pregnancy.
Collapse
Affiliation(s)
- Alexandr Ceasovschih
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- 2nd Internal Medicine Department, “Sf. Spiridon” Clinical Emergency Hospital, Iasi, Romania
| | - Victorița Șorodoc
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- 2nd Internal Medicine Department, “Sf. Spiridon” Clinical Emergency Hospital, Iasi, Romania
| | - Serghei Covantsev
- Department of Research and Clinical Development, Botkin Hospital, Moscow, Russia
| | - Anastasia Balta
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- 2nd Internal Medicine Department, “Sf. Spiridon” Clinical Emergency Hospital, Iasi, Romania
| | - Jamol Uzokov
- Department of Cardiology, Republican Specialized Scientific Practical Medical Center of Therapy and Medical Rehabilitation, Tashkent, Uzbekistan
| | - Sergio E Kaiser
- Discipline of Clinical and Experimental Pathophysiology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Abdallah Almaghraby
- Department of Cardiology, Ibrahim Bin Hamad Obaidallah Hospital, Ras Al Khaimah, United Arab Emirates
| | - Cătălina Lionte
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- 2nd Internal Medicine Department, “Sf. Spiridon” Clinical Emergency Hospital, Iasi, Romania
| | - Cristian Stătescu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- Department of Cardiology, “Prof. Dr. George I.M. Georgescu” Cardiovascular Diseases Institute, Iasi, Romania
| | - Radu A Sascău
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- Department of Cardiology, “Prof. Dr. George I.M. Georgescu” Cardiovascular Diseases Institute, Iasi, Romania
| | - Viviana Onofrei
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- Department of Cardiology, “Sf. Spiridon” Clinical Emergency Hospital, Iasi, Romania
| | - Raluca Ecaterina Haliga
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- 2nd Internal Medicine Department, “Sf. Spiridon” Clinical Emergency Hospital, Iasi, Romania
| | - Alexandra Stoica
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- 2nd Internal Medicine Department, “Sf. Spiridon” Clinical Emergency Hospital, Iasi, Romania
| | - Cristina Bologa
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- 2nd Internal Medicine Department, “Sf. Spiridon” Clinical Emergency Hospital, Iasi, Romania
| | - Ștefan Ailoaei
- Department of Cardiology, “Prof. Dr. George I.M. Georgescu” Cardiovascular Diseases Institute, Iasi, Romania
| | - Yusuf Ziya Şener
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkiye
| | - Nicholas G Kounis
- Department of Internal Medicine, Division of Cardiology, University of Patras Medical School, Patras, Greece
| | - Laurențiu Șorodoc
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- 2nd Internal Medicine Department, “Sf. Spiridon” Clinical Emergency Hospital, Iasi, Romania
| |
Collapse
|
49
|
Shukla AK, Awasthi K, Usman K, Banerjee M. Role of renin-angiotensin system/angiotensin converting enzyme-2 mechanism and enhanced COVID-19 susceptibility in type 2 diabetes mellitus. World J Diabetes 2024; 15:606-622. [PMID: 38680697 PMCID: PMC11045416 DOI: 10.4239/wjd.v15.i4.606] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/22/2024] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a disease that caused a global pandemic and is caused by infection of severe acute respiratory syndrome coronavirus 2 virus. It has affected over 768 million people worldwide, resulting in approximately 6900000 deaths. High-risk groups, identified by the Centers for Disease Control and Prevention, include individuals with conditions like type 2 diabetes mellitus (T2DM), obesity, chronic lung disease, serious heart conditions, and chronic kidney disease. Research indicates that those with T2DM face a heightened susceptibility to COVID-19 and increased mortality compared to non-diabetic individuals. Examining the renin-angiotensin system (RAS), a vital regulator of blood pressure and pulmonary stability, reveals the significance of the angiotensin-converting enzyme (ACE) and ACE2 enzymes. ACE converts angiotensin-I to the vasoconstrictor angiotensin-II, while ACE2 counters this by converting angiotensin-II to angiotensin 1-7, a vasodilator. Reduced ACE2 expression, common in diabetes, intensifies RAS activity, contributing to conditions like inflammation and fibrosis. Although ACE inhibitors and angiotensin receptor blockers can be therapeutically beneficial by increasing ACE2 levels, concerns arise regarding the potential elevation of ACE2 receptors on cell membranes, potentially facilitating COVID-19 entry. This review explored the role of the RAS/ACE2 mechanism in amplifying severe acute respiratory syndrome coronavirus 2 infection and associated complications in T2DM. Potential treatment strategies, including recombinant human ACE2 therapy, broad-spectrum antiviral drugs, and epigenetic signature detection, are discussed as promising avenues in the battle against this pandemic.
Collapse
Affiliation(s)
- Ashwin Kumar Shukla
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| | - Komal Awasthi
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| | - Kauser Usman
- Department of Medicine, King Georges’ Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Monisha Banerjee
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
- Institute of Advanced Molecular Genetics, and Infectious Diseases (IAMGID), University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| |
Collapse
|
50
|
Gao F, Lin W, Wang X, Liao M, Zhang M, Qin N, Chen X, Xia L, Chen Q, Sha O. Identification of receptors and factors associated with human coronaviruses in the oral cavity using single-cell RNA sequencing. Heliyon 2024; 10:e28280. [PMID: 38560173 PMCID: PMC10981076 DOI: 10.1016/j.heliyon.2024.e28280] [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: 10/24/2023] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) ravaged the world, and Coronavirus Disease 2019 (COVID-19) exhibited highly prevalent oral symptoms that had significantly impacted the lives of affected patients. However, the involvement of four human coronavirus (HCoVs), namely SARS-CoV-2, SARS-CoV, MERS-CoV, and HCoV-229E, in oral cavity infections remained poorly understood. We integrated single-cell RNA sequencing (scRNA-seq) data of seven human oral tissues through consistent normalization procedure, including minor salivary gland (MSG), parotid gland (PG), tongue, gingiva, buccal, periodontium and pulp. The Seurat, scDblFinder, Harmony, SingleR, Ucell and scCancer packages were comprehensively used for analysis. We identified specific cell clusters and generated expression profiles of SARS-CoV-2 and coronavirus-associated receptors and factors (SCARFs) in seven oral regions, providing direction for predicting the tropism of four HCoVs for oral tissues, as well as for dental clinical treatment. Based on our analysis, it appears that various SCARFs, including ACE2, ASGR1, KREMEN1, DPP4, ANPEP, CD209, CLEC4G/M, TMPRSS family proteins (including TMPRSS2, TMPRSS4, and TMPRSS11A), and FURIN, are expressed at low levels in the oral cavity. Conversely, BSG, CTSB, and CTSL exhibit enrichment in oral tissues. Our study also demonstrates widespread expression of restriction factors, particularly IFITM1-3 and LY6E, in oral cells. Additionally, some replication, assembly, and trafficking factors appear to exhibit broad oral tissues expression patterns. Overall, the oral cavity could potentially serve as a high-risk site for SARS-CoV-2 infection, while displaying a comparatively lower degree of susceptibility towards other HCoVs (including SARS-CoV, MERS-CoV and HCoV-229E). Specifically, MSG, tongue, and gingiva represent potential sites of vulnerability for four HCoVs infection, with the MSG exhibiting a particularly high susceptibility. However, the expression patterns of SCARFs in other oral sites demonstrate relatively intricate and may only be specifically associated with SARS-CoV-2 infection. Our study sheds light on the mechanisms of HCoVs infection in the oral cavity as well as gains insight into the characteristics and distribution of possible HCoVs target cells in oral tissues, providing potential therapeutic targets for HCoVs infection in the oral cavity.
Collapse
Affiliation(s)
- Feng Gao
- School of Dentistry, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Institute of Dental Research, Shenzhen University, Shenzhen, China
| | - Weiming Lin
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xia Wang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- The Chinese University of Hong Kong Shenzhen, School of Medicine, Shenzhen, China
| | - Mingfeng Liao
- The Third People's Hospital of Shenzhen, Shenzhen, China
| | - Mingxia Zhang
- The Third People's Hospital of Shenzhen, Shenzhen, China
| | - Nianhong Qin
- Department of Stomatology, Shenzhen People's Hospital, Shenzhen, China
| | - Xianxiong Chen
- School of Dentistry, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Lixin Xia
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Ou Sha
- School of Dentistry, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Institute of Dental Research, Shenzhen University, Shenzhen, China
| |
Collapse
|