1
|
Kasem Ali Sliman R, Cohen H, Shehadeh S, Batcir R, Alter YE, Cohen K, Koren I, Halabi I, Sliman H, Saied MH. Pediatric autoimmune diseases in the light of COVID-19 pandemic, A retrospective observational big data study. J Transl Autoimmun 2025; 10:100281. [PMID: 40162434 PMCID: PMC11951201 DOI: 10.1016/j.jtauto.2025.100281] [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: 12/21/2024] [Revised: 02/18/2025] [Accepted: 03/01/2025] [Indexed: 04/02/2025] Open
Abstract
Background The COVID-19 pandemic has raised concerns about potential links between SARS-CoV-2 infection and autoimmune diseases. This study investigated changes in the incidence rate (IR) of autoimmune diseases among children following the pandemic's onset. Methods A retrospective cross-sectional study analyzed data from Clalit Health Services, Israel's largest healthcare provider, examining the IR of different autoimmune diseases in children aged 0-18. The study compared pre-pandemic (2019) with pandemic/post-pandemic periods (2020-2023), encompassing a cohort of over 1.5 million children. Results Significant IR increases were observed across multiple autoimmune diseases. Rheumatic diseases (Juvenile Idiopathic Arthritis, Systemic Lupus Erythematosus, Henoch Schoenlein Purpura (HSP)) showed consistent increases, with HSP demonstrating the most pronounced trend. Endocrine disorders exhibited diverse patterns, with autoimmune thyroid diseases and Type 1 diabetes showing overall increases, while diabetic ketoacidosis exhibited an initial spike followed by a decline. Gastrointestinal diseases displayed heterogeneous patterns; Celiac disease and Ulcerative colitis showed general increases, Crohn's disease showed a downward trend, and autoimmune hepatitis exhibited an initial significant decrease followed by a significant increase. Dermatological conditions, including Psoriasis and Vitiligo, demonstrated consistent elevations throughout 2020-2023. Immune Thrombocytopenia Purpura showed initial decreases followed by significant increases in 2022-2023. Conclusions This comprehensive analysis reveals significant changes in pediatric autoimmune disease incidence following the COVID-19 pandemic, suggesting potential associations between SARS-CoV-2 infection and autoimmune dysregulation. The diverse patterns observed across different conditions highlight the complex interplay between viral infection and autoimmunity, emphasizing the need for continued surveillance and investigation of long-term immunological consequences of COVID-19 in pediatric populations.
Collapse
Affiliation(s)
- Rim Kasem Ali Sliman
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
| | - Hilla Cohen
- Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
| | - Shereen Shehadeh
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Infectious Disease Unit, Carmel Medical Center, Haifa, Israel
| | - Reut Batcir
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Gastroenterology Unit, Carmel Medical Center, Haifa, Israel
| | - Yigal Elenberg Alter
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Gastroenterology Unit, Carmel Medical Center, Haifa, Israel
| | - Keren Cohen
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Ilana Koren
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Inbal Halabi
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Hussein Sliman
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Cardiology, Carmel Medical Center, Heart Center, Haifa, Israel
| | - Mohamad Hamad Saied
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital/University Medical Center, Utrecht, the Netherlands
| |
Collapse
|
2
|
Adhikari P, Jawad B, Ching WY. Electronic Interactions Between the Receptor-Binding Domain of Omicron Variants and Angiotensin-Converting Enzyme 2: A Novel Amino Acid-Amino Acid Bond Pair Concept. Molecules 2025; 30:2061. [PMID: 40363865 PMCID: PMC12073306 DOI: 10.3390/molecules30092061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/29/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
SARS-CoV-2 remains a severe threat to worldwide public health, particularly as the virus continues to evolve and diversify into variants of concern (VOCs). Among these VOCs, Omicron variants exhibit unique phenotypic traits, such as immune evasion, transmissibility, and severity, due to numerous spike protein mutations and the rapid subvariant evolution. These Omicron subvariants have more than 15 mutations in the receptor-binding domain (RBD), a region of the SARS-CoV-2 spike protein that is important for recognition and binding with the angiotensin-converting enzyme 2 (ACE2) human receptor. To address the impact of these high numbers of Omicron mutations on the binding process, we have developed a novel method to precisely quantify amino acid interactions via the amino acid-amino acid bond pair (AABP). We applied this concept to investigate the interface interactions of the RBD-ACE2 complex in four Omicron Variants (BA.1, BA.2, BA.5, and XBB.1.16) with its Wild Type counterpart. Based on the AABP analysis, we have identified all the sites that are affected by mutation and have provided evidence that unmutated sites are also impacted by mutation. We have calculated that the binding between RBD and ACE2 is strongest in OV BA.1, followed by OV BA.2, WT, OV BA.5, and OV XBB.1.16. We also present the partial charge values for all 311 residues across these five models. Our analysis provides a detailed understanding of changes caused by mutation in each Omicron interface complex.
Collapse
Affiliation(s)
- Puja Adhikari
- Department of Physics and Astronomy, University of Missouri-Kansas City, Kansas City, MO 64110, USA; (B.J.); (W.-Y.C.)
| | - Bahaa Jawad
- Department of Physics and Astronomy, University of Missouri-Kansas City, Kansas City, MO 64110, USA; (B.J.); (W.-Y.C.)
- College of Applied Sciences, University of Technology, Baghdad 10066, Iraq
| | - Wai-Yim Ching
- Department of Physics and Astronomy, University of Missouri-Kansas City, Kansas City, MO 64110, USA; (B.J.); (W.-Y.C.)
| |
Collapse
|
3
|
da Silva FPG, Matte R, Wiedmer DB, da Silva APG, Menin RM, Barbosa FB, Meneguzzi TAM, Pereira SB, Fausto AT, Klug L, Melim BP, Beltrão CJ. HIF-1α Pathway in COVID-19: A Scoping Review of Its Modulation and Related Treatments. Int J Mol Sci 2025; 26:4202. [PMID: 40362439 PMCID: PMC12071378 DOI: 10.3390/ijms26094202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
The COVID-19 pandemic, driven by SARS-CoV-2, has led to a global health crisis, highlighting the virus's unique molecular mechanisms that distinguish it from other respiratory pathogens. It is known that the Hypoxia-Inducible Factor 1α (HIF-1α) activates a complex network of intracellular signaling pathways regulating cellular energy metabolism, angiogenesis, and cell survival, contributing to the wide range of clinical manifestations of COVID-19, including Post-Acute COVID-19 Syndrome (PACS). Emerging evidence suggests that dysregulation of HIF-1α is a key driver of systemic inflammation, silent hypoxia, and pathological tissue remodeling in both the acute and post-acute phases of the disease. This scoping review was conducted following PRISMA-ScR guidelines and registered in INPLASY. It involved a literature search in Scopus and PubMed, supplemented by manual reference screening, with study selection facilitated by Rayyan software. Our analysis clarifies the dual role of HIF-1α, which may either worsen inflammatory responses and viral persistence or support adaptive mechanisms that reduce cellular damage. The potential for targeting HIF-1α therapeutically in COVID-19 is complex, requiring further investigation to clarify its precise role and translational applications. This review deepens the molecular understanding of SARS-CoV-2-induced cellular and tissue dysfunction in hypoxia, offering insights for improving clinical management strategies and addressing long-term sequelae.
Collapse
Affiliation(s)
- Felipe Paes Gomes da Silva
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| | - Rafael Matte
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| | - David Batista Wiedmer
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| | - Arthur Paes Gomes da Silva
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| | - Rafaela Makiak Menin
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| | - Fernanda Bressianini Barbosa
- School of Medicine, Faculdade Evangélica Mackenzie do Paraná, R. Padre Anchieta, no. 2770—Bigorrilho, Curitiba 80730-000, PR, Brazil;
| | - Thainá Aymê Mocelin Meneguzzi
- School of Medicine, Universidade Nove de Julho—UNINOVE, Rua Vergueiro, 249—Liberdade, São Paulo 01504-001, SP, Brazil; (T.A.M.M.); (B.P.M.)
| | - Sabrina Barancelli Pereira
- School of Medicine, Universidade Positivo—UP, R. Professor Pedro Viriato Parigot de Souza, 5300, Curitiba 81280-330, PR, Brazil; (S.B.P.); (A.T.F.); (L.K.)
| | - Amanda Terres Fausto
- School of Medicine, Universidade Positivo—UP, R. Professor Pedro Viriato Parigot de Souza, 5300, Curitiba 81280-330, PR, Brazil; (S.B.P.); (A.T.F.); (L.K.)
| | - Larissa Klug
- School of Medicine, Universidade Positivo—UP, R. Professor Pedro Viriato Parigot de Souza, 5300, Curitiba 81280-330, PR, Brazil; (S.B.P.); (A.T.F.); (L.K.)
| | - Bruna Pinheiro Melim
- School of Medicine, Universidade Nove de Julho—UNINOVE, Rua Vergueiro, 249—Liberdade, São Paulo 01504-001, SP, Brazil; (T.A.M.M.); (B.P.M.)
| | - Claudio Jose Beltrão
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| |
Collapse
|
4
|
Hong P, Waldenberger M, Pritsch M, Gilberg L, Brand I, Bruger J, Frese J, Castelletti N, Garí M, Geldmacher C, Hoelscher M, Peters A, Matías-García PR. Differential DNA methylation 7 months after SARS-CoV-2 infection. Clin Epigenetics 2025; 17:60. [PMID: 40251596 PMCID: PMC12008906 DOI: 10.1186/s13148-025-01866-4] [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: 01/17/2025] [Accepted: 03/26/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), and SARS-CoV-2 has been linked to changes in DNA methylation (DNAm) patterns. Studies focused on post-SARS-CoV-2 infection and DNAm have been mainly carried out among severe COVID-19 cases or without distinguishing the severity of cases. However, investigations into mild and asymptomatic cases after SARS-CoV-2 infection are limited. In this study, we analyzed DNAm patterns of mild and asymptomatic cases seven months after SARS-CoV-2 infection in a household setting by conducting epigenome-wide association studies (EWAS). RESULTS We identified DNAm changes at 42 CpG sites associated with anti-SARS-CoV-2 antibody levels. We additionally report EWAS between COVID-19 cases and controls, with the case status being confirmed by either an antibody test or a PCR test. The EWAS with an antibody test case definition identified 172 CpG sites to be differentially methylated, while the EWAS with a PCR test case definition identified 502 CpG sites. Two common sites, namely cg17126990 (annotated to AFAP1L2) and cg25483596 (annotated to PC), were identified to be hypermethylated across the three EWAS. Both CpG sites have been reported to be involved in molecular pathways after SARS-CoV-2 infection. While AFAP1L2 has been found to be upregulated after SARS-CoV-2 infection, the pyruvate carboxylase (PC) activity seems to be affected by SARS-CoV-2 infection resulting in changes to the host cell metabolism. Additionally, an EWAS to assess persistent health restrictions among PCR-confirmed cases showed 40 CpG sites to be differentially methylated. CONCLUSIONS We detected associations between DNAm in individuals who had asymptomatic and mild SARS-CoV-2 infections as compared to their household controls. These findings contribute to our understanding of the molecular consequences of SARS-CoV-2 infection observed months after infection.
Collapse
Grants
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- European Union’s Horizon 2020 research and innovation program, ORCHESTRA
- Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) (4209)
Collapse
Affiliation(s)
- Peizhen Hong
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany.
- Pettenkofer School of Public Health, Munich, Germany.
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Michael Pritsch
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Leonard Gilberg
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Infectious Diseases, LMU University Hospital, LMU Munich, Munich, Germany
| | - Isabel Brand
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU, Munich, Germany
| | - Jan Bruger
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Jonathan Frese
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Noemi Castelletti
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mercè Garí
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christof Geldmacher
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Immunology, Infection and Pandemic Research, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 80799, Munich, Germany
| | - Michael Hoelscher
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Center for International Health (CIH), University Hospital, LMU Munich, Munich, Germany
- Immunology, Infection and Pandemic Research, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 80799, Munich, Germany
| | - Annette Peters
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Pettenkofer School of Public Health, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Pamela R Matías-García
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
5
|
Aderinto N, Olatunji G, Kokori E, Ogieuhi IJ, Yusuf IA, Egbunu E, Ukoaka BM, Babalola AE, Adefusi TO, Aboje JE, Moradeyo A. COVID-19 and cognitive impairment: a review of the emerging evidence. DISCOVER MENTAL HEALTH 2025; 5:56. [PMID: 40244315 PMCID: PMC12006571 DOI: 10.1007/s44192-025-00189-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 04/10/2025] [Indexed: 04/18/2025]
Abstract
The recent surge of COVID-19 cases has raised concerns about its potential long-term effects on cognitive function. This review explores the growing body of research investigating the link between COVID-19 infection and cognitive impairment. Studies employing observational, longitudinal, and case-control designs reveal a concerning prevalence of cognitive impairment in survivors, affecting domains like attention, memory, executive function, and processing speed. The persistence of these deficits for months after the initial infection highlights the potential for long-term consequences. While the precise mechanisms remain under investigation, potential contributing factors include neuroinflammation, hypoxia, and psychological effects. Limitations within the current research landscape necessitate further investigation into the long-term trajectory of cognitive decline, the potential for intervention and recovery, and the role of vaccination in mitigating these effects. Understanding the multifaceted nature of this issue is crucial for developing effective strategies to ensure optimal cognitive health outcomes for COVID-19 survivors.
Collapse
Affiliation(s)
- Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Nigeria.
| | - Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | | | | | | | | | | | - John Ehi Aboje
- College of Health Sciences, Benue State University, Makurdi, Benue, Nigeria
| | - Abdulrahmon Moradeyo
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| |
Collapse
|
6
|
Noor MS, Ferdous S, Salehi R, Gates H, Dey S, Raghunath VS, Zargar MR, Chowdhury R. Next-generation metabolic models informed by biomolecular simulations. Curr Opin Biotechnol 2025; 92:103259. [PMID: 39827498 DOI: 10.1016/j.copbio.2025.103259] [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: 11/18/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025]
Abstract
Metabolic modeling is essential for understanding the mechanistic bases of cellular metabolism in various organisms, from microbes to humans, and the design of fitter microbial strains. Metabolic networks focus on the overall fluxes through biochemical reactions that implicitly rely on several biochemical processes, such as active or diffusive uptake (or export) of nutrients (or metabolites), enzymatic turnover of metabolites, and metal-cofactor enzyme interactions. Despite independent progress in biomolecular simulations, they have yet to be integrated to inform metabolic models. We explore the evolution of computational metabolic modeling approaches, starting with flux balance analysis, dynamic, kinetic delineations of metabolic shifts in single organisms within cells and across tissues, and mutually informing, community-level modeling frameworks and provide a narrative to tie in biomolecular simulations and machine learning predictions to usher the new phase of structure-guided synthetic biology applications. These additions and prospective novel ones are likely to open hitherto untapped paradigms for optimizing/understanding metabolic pathways toward improving bioproduction of protein and small molecule products with downstream applications in health, environment, energy, and sustainability.
Collapse
Affiliation(s)
- Mohammed S Noor
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Sakib Ferdous
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Rahil Salehi
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Hannah Gates
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Supantha Dey
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Vaishnavey S Raghunath
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Mohammad R Zargar
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Ratul Chowdhury
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA.
| |
Collapse
|
7
|
Kruger A, Joffe D, Lloyd-Jones G, Khan MA, Šalamon Š, Laubscher GJ, Putrino D, Kell DB, Pretorius E. Vascular Pathogenesis in Acute and Long COVID: Current Insights and Therapeutic Outlook. Semin Thromb Hemost 2025; 51:256-271. [PMID: 39348850 PMCID: PMC11906225 DOI: 10.1055/s-0044-1790603] [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: 10/02/2024]
Abstract
Long coronavirus disease 2019 (COVID-19)-a postacute consequence of severe acute respiratory syndrome coronavirus 2 infection-manifests with a broad spectrum of relapsing and remitting or persistent symptoms as well as varied levels of organ damage, which may be asymptomatic or present as acute events such as heart attacks or strokes and recurrent infections, hinting at complex underlying pathogenic mechanisms. Central to these symptoms is vascular dysfunction rooted in thrombotic endothelialitis. We review the scientific evidence that widespread endothelial dysfunction (ED) leads to chronic symptomatology. We briefly examine the molecular pathways contributing to endothelial pathology and provide a detailed analysis of how these cellular processes underpin the clinical picture. Noninvasive diagnostic techniques, such as flow-mediated dilation and peripheral arterial tonometry, are evaluated for their utility in identifying ED. We then explore mechanistic, cellular-targeted therapeutic interventions for their potential in treating ED. Overall, we emphasize the critical role of cellular health in managing Long COVID and highlight the need for early intervention to prevent long-term vascular and cellular dysfunction.
Collapse
Affiliation(s)
- Arneaux Kruger
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - David Joffe
- Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney, Australia
- World Health Network, Cambridge, Massachusetts
| | - Graham Lloyd-Jones
- Department of Radiology, Salisbury District Hospital, Salisbury NHS Foundation Trust, United Kingdom
| | - Muhammed Asad Khan
- World Health Network, Cambridge, Massachusetts
- Directorate of Respiratory Medicine, Manchester University Hospitals, Wythenshawe Hospital, Manchester, United Kingdom
| | | | | | - David Putrino
- Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney, Australia
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- World Health Network, Cambridge, Massachusetts
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
8
|
Joo HA, Kang BC, Kim TS, Kang WS, Park HJ, Chung JW, Ahn JH. A trend of otologic diseases during the coronavirus disease 2019 pandemic period. Acta Otolaryngol 2025; 145:277-282. [PMID: 39898862 DOI: 10.1080/00016489.2025.2459343] [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: 12/10/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) changed the practice of medicine, and various anti-contagion restrictions were implemented worldwide. OBJECTIVES This study aimed to evaluate the annual trend in the incidence of otologic diseases during the COVID-19 pandemic and determine the influence of social restrictions on these diseases. MATERIALS AND METHODS We retrospectively reviewed the number of patients treated for otologic diseases, particularly those associated with infectious pathophysiology, and the total number of patients in the otolaryngology departments at three tertiary referral centers in South Korea. RESULTS The number of patients who underwent ventilation tube insertion due to persistent otitis media with effusion (OME) significantly decreased (raw number: -37.1%, incidence: -2.04‰) after the onset of the COVID-19 pandemic. However, a sharp increase was observed in 2023 (raw number: +42.9%, incidence: +1.63‰) when the pandemic officially ended. The number of patients newly diagnosed with sudden sensorineural hearing loss (SSNHL) gradually increased during the pandemic period. CONCLUSIONS AND SIGNIFICANCE Restrictive measures to control COVID-19 spread positively influenced the reduction in the incidence of OME. Whether COVID-19 is a direct risk factor for SSNHL remains uncertain, but the potential impact of the virus itself or the COVID-19 vaccine on the auditory system appears to exist.
Collapse
Affiliation(s)
- Hye Ah Joo
- Department of Otorhinolaryngology-Head and Neck Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Byung Chul Kang
- Department of Otorhinolaryngology-Head and Neck Surgery, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Tae Su Kim
- Department of Otolaryngology, Kangwon National University Hospital, Chuncheon, Korea
| | - Woo Seok Kang
- Department of Otorhinolaryngology-Head and Neck Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hong Ju Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jong Woo Chung
- Department of Otorhinolaryngology-Head and Neck Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Joong Ho Ahn
- Department of Otorhinolaryngology-Head and Neck Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Boira I, Chiner E. Sleep and Respiratory Infections. Semin Respir Crit Care Med 2025. [PMID: 39900109 DOI: 10.1055/a-2531-1018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Sleep disorders that involve circadian rhythm disruption and sleep-disordered breathing (SDB) such as obstructive sleep apnea (OSA) are closely linked to respiratory infections. SDB leads to a proinflammatory state due to intermittent hypoxia, sleep fragmentation, increased oxidative stress, and elevation of inflammatory mediators such as tumor necrosis factor (TNF), interleukin-6 (IL-6), and C-reactive protein (CRP). Furthermore, inflammatory mediator levels correlate with SDB severity, especially in people with OSA. Nocturnal microaspiration, gastroesophageal reflux, and associated comorbidities (e.g., obesity) increase the risk of community-acquired pneumonia, viral infections such as SARS-CoV-2, respiratory complications, and death. OSA has been associated with post-COVID syndrome. It also increases the risk of postoperative complications in both adults and children. Circadian rhythm disorders such as insomnia predispose to immune disorders and increase the risk of infection. Chronic conditions such as bronchiectasis, with or without concomitant cystic fibrosis, can lead to structural sleep changes and increase the risk of OSA due to chronic cough, arousals, aspirations, hypoxia, upper airway edema, and overexpression of proinflammatory cytokines. The protective effect of treatment for sleep disorders against respiratory infection is currently unknown. However, in people presenting with respiratory infection, it is important to test for SDB to prevent complications.
Collapse
Affiliation(s)
- Ignacio Boira
- Sleep Unit, Pneumology Department, San Juan de Alicante University Hospital, Alicante, Spain
| | - Eusebi Chiner
- Sleep Unit, Pneumology Department, San Juan de Alicante University Hospital, Alicante, Spain
| |
Collapse
|
10
|
Schnaubelt S, Jakobljevich A, Brock R, Oppenauer J, Kornfehl A, Eibensteiner F, Veigl C, Perkmann T, Haslacher H, Strassl R, Reindl-Schwaighofer R, Schlager O, Sulzgruber P. The Relation of Angiotensin-Converting Enzyme 2, Renin-Angiotensin-Aldosterone System Inhibitors, and Arterial Stiffness in Acute COVID-19 Emergency Department Patients-A Prospective Observational Study. J Clin Med 2025; 14:2233. [PMID: 40217682 PMCID: PMC11989675 DOI: 10.3390/jcm14072233] [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: 02/12/2025] [Revised: 03/13/2025] [Accepted: 03/22/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causing coronavirus disease 2019 (COVID-19) can damage the endothelium and increase arterial stiffness, potentially leading to adverse cardiovascular events. In parallel, systemic inflammation in COVID-19 also impacts endothelial function. Angiotensin-converting enzyme 2 (ACE2) promotes vasodilation and anti-inflammatory effects, but also facilitates SARS-CoV-2 entry into human cells. Thus, concerns have been raised about the use of RAAS inhibitors (RAASi) in COVID-19 patients due to potential ACE2 upregulation. However, the clinical significance of increased plasma ACE2 (sACE2) in RAASi-treated COVID-19 patients remains unclear. Methods: This prospective, single-centre study evaluated RAASi, sACE2, and vascular function in acutely ill patients with COVID-19 in comparison with acutely ill patients without COVID-19. Adult emergency department patients with confirmed or suspected COVID-19 were enrolled and underwent pulse wave velocity, ankle brachial index, and sACE2 measurements. Results: In the 152 included patients (50% female, median age 62 years, 68% COVID-19 positive), the sACE2 values were slightly higher in the COVID-19 (0.485 [0.364-1.329]) than in the non-COVID-19 subgroup (0.458 [0.356-1.138]; p = 0.70). No significant differences in sACE2 were observed between patients with and without RAASi, regardless of COVID-19 status. Pulse wave velocity values differed significantly between groups (p = 0.015). Conclusions: In emergency department patients, sACE2 was upregulated in COVID-19 patients, probably due to oxidative stress and inflammation. RAASi did not increase sACE2, but may have protective effects against inflammation. Elevated sACE2 appeared to have a beneficial effect on arterial stiffness in all patients. These findings support continued RAASi therapy in COVID-19 patients to protect against chronic inflammation and apoptosis.
Collapse
Affiliation(s)
- Sebastian Schnaubelt
- Department of Emergency Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Emergency Medical Service Vienna, 1030 Vienna, Austria
| | - Anna Jakobljevich
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | - Roman Brock
- Department of Emergency Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Julia Oppenauer
- Department of Emergency Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Andrea Kornfehl
- Department of Emergency Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Felix Eibensteiner
- Department of Emergency Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Christoph Veigl
- Department of Emergency Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Perkmann
- Department Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Helmuth Haslacher
- Department Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Robert Strassl
- Division of Clinical Virology, Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Roman Reindl-Schwaighofer
- Division of Nephrology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Oliver Schlager
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Patrick Sulzgruber
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
11
|
Barreto Fernandes TF, Pilotto JH, Cezar PA, Côrtes FH, Morgado MG, Giacoia-Gripp CBW, De Sá NBR, Da Silva Cazote A, Neves AF, Quintana MDSB, Diniz Ribeiro MP, Cardoso SW, Veloso VG, Grinsztejn B, De Almeida DV. Modulation of RAAS receptors and miRNAs in COVID-19: implications for disease severity, immune response, and potential therapeutic targets. BMC Infect Dis 2025; 25:399. [PMID: 40128651 PMCID: PMC11934810 DOI: 10.1186/s12879-025-10803-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
The SARS-CoV-2 spike protein interacts with ACE2, a key receptor within the renin-angiotensin-aldosterone system (RAAS), which plays a critical role in maintaining vascular homeostasis, regulating blood pressure, and modulating inflammation. An observational study analyzed the gene expression profiles of RAAS receptors and associated miRNAs in 88 hospitalized COVID-19 patients and 20 healthy controls, comparing the acute and post-acute phases to assess their impact on disease severity and recovery. Our findings revealed an association between reduced MAS1 expression in both advanced age (P = 0.03) and the need for oxygen supplementation (P = 0.04). Additionally, reduced ACE expression was associated with worse mortality outcomes (P = 0.01). Notably, ACE2 and TMPRSS2 expression was significantly decreased (P < 0.0001) in individuals requiring oxygen supplementation and in those with diabetes mellitus during both the acute and post-COVID-19 phases, further highlighting the impact of these conditions on RAAS. The miRNA analysis revealed significant downregulation of miR-200c (P = 0.005), miR-let-7 (P = 0.01), and miR-122 (P = 0.03) in acute-phase COVID-19 patients. This dysregulation contributes to the inflammatory response and highlights the interaction between viral entry and immune regulation. These results underscore the significance of the ACE2/Ang-(1-7)/MAS1 axis in inflammation regulation and suggest that targeting this pathway may have therapeutic potential. Our study provides valuable insights into the molecular mechanisms of COVID-19 pathogenesis and identifies the modulation of RAAS receptors and miRNAs as promising biomarkers for disease severity and potential therapeutic interventions. CLINICAL TRIAL: Not applicable.
Collapse
Affiliation(s)
| | - Jose Henrique Pilotto
- Laboratório de AIDS e Imunologia Molecular, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brasil
| | - Priscila Alves Cezar
- Laboratório de AIDS e Imunologia Molecular, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brasil
| | - Fernanda Heloise Côrtes
- Laboratório de AIDS e Imunologia Molecular, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brasil
| | - Mariza G Morgado
- Laboratório de AIDS e Imunologia Molecular, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brasil
| | | | | | - Andressa Da Silva Cazote
- Laboratório de AIDS e Imunologia Molecular, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brasil
| | - Agatha Freixinho Neves
- Laboratório de AIDS e Imunologia Molecular, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brasil
| | | | | | | | - Valdiléa G Veloso
- Instituto Nacional de Infectologia Evandro Chagas, FIOCRUZ, Rio de Janeiro, Brasil
| | - Beatriz Grinsztejn
- Instituto Nacional de Infectologia Evandro Chagas, FIOCRUZ, Rio de Janeiro, Brasil
| | | |
Collapse
|
12
|
Scalzo PL, Marshall AG, Soriano S, Curry K, Dulay M, Hodics T, Quigley EMM, Treangen TJ, Piskorz MM, Villapol S. Gut Microbiome dysbiosis and immune activation correlate with somatic and neuropsychiatric symptoms in COVID-19 patients. J Transl Med 2025; 23:327. [PMID: 40087795 PMCID: PMC11907868 DOI: 10.1186/s12967-025-06348-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Infection with SARS-CoV-2, the virus responsible for COVID-19, can lead to a range of physical symptoms and mental health challenges, including stress, anxiety, and depression. These effects are particularly pronounced in hospitalized patients, likely due to the virus's direct and indirect impact on the nervous system. Gut dysbiosis, an imbalance in the gut microbiome, has been implicated in immune dysfunction and chronic inflammation in COVID-19 patients. However, the interactions between gut microbiome composition and the physical and mental symptoms of COVID-19 remain incompletely understood. METHODS We investigated the association between physical and mental symptoms, cytokine profiles, and gut microbiota composition in 124 hospitalized COVID-19 patients. We collected data on demographics, COVID-19 severity, and mental health indicators (stress, anxiety, and depression). Gut microbiome profiling was performed using full-length 16 S rRNA gene sequencing to evaluate microbial diversity and composition. RESULTS COVID-19 severity was categorized as low (27.4%), moderate (29.8%), or critical (42.8%). Common symptoms included fever (66.1%) and cough (55.6%), while somatic symptoms (27.3%), anxiety (27.3%), depressive symptoms (39%), and stress (80.5%) were frequently self-reported. Elevated interleukin-6 levels in severe cases highlighted systemic inflammation, reduced gut bacterial diversity, particularly among women and obese patients, correlated with higher disease severity. Notably, the genus Mitsuokella was associated with increased physical symptoms and mental distress, while Granulicatella was linked to critical illness. CONCLUSIONS Our findings reveal significant associations between mental health status, systemic inflammation, and gut dysbiosis in hospitalized COVID-19 patients. These results indicate the potential for microbiome-targeted therapies to mitigate psychological and physical complications and improve recovery outcomes in this population.
Collapse
Affiliation(s)
- Paula L Scalzo
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Austin G Marshall
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Computer Science, Rice University, Houston, TX, USA
| | - Sirena Soriano
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Kristen Curry
- Department of Computer Science, Rice University, Houston, TX, USA
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Mario Dulay
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Timea Hodics
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Eamonn M M Quigley
- Lynda K. and David M. Underwood Center for Digestive Health, Houston Methodist Hospital, Houston, TX, USA
| | - Todd J Treangen
- Department of Computer Science, Rice University, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - María M Piskorz
- Department of Neurogastroenterology, Hospital de Clinicas José de San Martin, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sonia Villapol
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA.
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Neuroscience in Neurological Surgery, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
13
|
Lin MW, Lin CH, Chang JR, Chiang HH, Wu TH, Lin CS. The influence of PM2.5 exposure on SARS-CoV-2 infection via modulating the expression of angiotensin converting enzyme II. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136887. [PMID: 39700942 DOI: 10.1016/j.jhazmat.2024.136887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Particulate matter 2.5 (PM2.5) pollution and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic are the greatest environmental health issues worldwide. Several statistics revealed the significant positive correlation between the morbidity of coronavirus disease-19 (COVID-19) and the levels of air pollution. Nevertheless, there is no direct experimental evidence to indicate the effect of PM2.5 exposure on SARS-CoV-2 infection. The objective of this study was to evaluate whether the infection of SARS-CoV-2 affected by PM2.5 through angiotensin-converting enzyme II (ACE2) expression enhances and investigate the function of ACE2 in lung injury induced by PM2.5. An animal model of PM2.5-induced lung injury was established using wild-type (WT, C57BL/6), human ACE2 transgenic (K18-hACE2 TG), and murine ACE2 gene knockout (mACE2 KO) mice. The results indicate that PM2.5 exposure facilitates SARS-CoV-2 infection through inducing ACE2 expression in vitro (10 μg/mL) and in vivo (6.25 mg/kg/day in 50 μL saline). The levels of ACE, inflammatory cytokines, and mitogen-activated protein kinase (MAPK) proteins in WT, K18-hACE TG and mACE2 KO mice were significantly increased after PM2.5 instillation. The severest PM2.5-induced lung damage was observed in mACE2 KO mice. In summary, ACE2 plays a double-edged sword role in lung injury, PM2.5 exposure contributed to SARS-CoV-2 infection through inducing ACE2 expression, but ACE2 also protected pulmonary inflammation from PM2.5 challenge.
Collapse
Affiliation(s)
- Meng-Wei Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Cheng-Han Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Jia-Rong Chang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Hua-Hsin Chiang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Ting-Hsuan Wu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Chih-Sheng Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| |
Collapse
|
14
|
Abou Mansour M, El Rassi C, Sleem B, Borghol R, Arabi M. Thromboembolic Events in the Era of COVID-19: A Detailed Narrative Review. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2025; 2025:3804576. [PMID: 40226433 PMCID: PMC11986918 DOI: 10.1155/cjid/3804576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/14/2025] [Indexed: 04/15/2025]
Abstract
COVID-19, caused by the SARS-CoV-2 virus, is not only characterized by respiratory symptoms but is also associated with a wide range of systemic complications, including significant hematologic abnormalities. This is a comprehensive review of the current literature, using PubMed and Google Scholar, on the pathophysiology and incidence of thromboembolic events in COVID-19 patients and thromboprophylaxis. COVID-19 infection induces a prothrombotic state in patients through the dysregulation of the renin-angiotensin-aldosterone system (RAAS), endothelial dysfunction, elevated von Willebrand factor (vWF), and a dysregulated immune response involving the complement system and neutrophil extracellular traps (NETs). As a result, thromboembolic complications have emerged in COVID-19 cases, occurring more frequently in severe cases and hospitalized patients. These thrombotic events affect both venous and arterial circulation, with increased incidences of deep venous thrombosis (DVT), pulmonary embolism (PE), systemic arterial thrombosis, and myocardial infarction (MI). While DVT and PE are more common, the literature highlights the potential lethal consequences of arterial thromboembolism (ATE). This review also briefly examines the ongoing discussions regarding the use of anticoagulants for the prevention of thrombotic events in COVID-19 patients. While theoretically promising, current studies have yielded varied outcomes: Some suggest potential benefits, whereas others report an increased risk of bleeding events among hospitalized patients. Therefore, further large-scale studies are needed to assess the efficacy and safety of anticoagulants for thromboprophylaxis in COVID-19 patients.
Collapse
Affiliation(s)
- Maria Abou Mansour
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Christophe El Rassi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Bshara Sleem
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Raphah Borghol
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Hematology-Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mariam Arabi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Cardiology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
15
|
Bostancı Ö, Karaduman E, Yılmaz AK, Kabadayı M, Bilgiç S. Midterm Effects of SARS-CoV-2 on Respiratory Function in Judokas With and Without Exercise-Induced Bronchoconstriction: A Retrospective Study. Clin J Sport Med 2025; 35:162-168. [PMID: 39626059 DOI: 10.1097/jsm.0000000000001312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 11/07/2023] [Indexed: 01/04/2025]
Abstract
OBJECTIVES The clinical consequences of coronavirus infection in elite judokas with exercise-induced bronchoconstriction (EIB) are unclear. We aimed to determine potential respiratory function abnormalities and recovery in athletes with and without EIB after severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. DESIGN Retrospective cohort study. SETTING Türkiye Olympic Preparation Centre. PARTICIPANTS This retrospective study analyzed data collected from 25 consecutive elite judokas diagnosed with and without EIB and SARS-CoV-2 infection, routinely followed at an Olympic Sports Center between September 2020 and 2021. INDEPENDENT VARIABLES Respiratory muscle strength and pulmonary function data were collected before and up to 90 days after SARS-CoV-2 infection. MAIN OUTCOME MEASURES Measurements included maximal inspiratory pressure (MIP), maximal expiratory pressure (MEP), forced expiratory volume in 1 second (FEV 1 ), forced vital capacity (FVC), FEV 1 /FVC ratio, and peak expiratory flow (PEF). RESULTS Infected athletes with EIB had more markedly reduced respiratory muscle strength and pulmonary function than those without EIB. Maximal inspiratory pressure was decreased by 14% and MEP by 8% from baseline in infected athletes with EIB during follow-up. Likewise, FEV 1 and FVC decreased by 4%. Maximal inspiratory pressure, MEP, FEV 1 , and FVC remained abnormal after 90 days of SARS-CoV-2 infection in EIB athletes but normalized rapidly in non-EIB athletes. Peak expiratory flow seemed unaffected during follow-up. Exercise-induced bronchoconstriction severity was moderately correlated with the maximum fall in MEP during follow-up. CONCLUSIONS Severe acute respiratory syndrome coronavirus-2 infection notably decreases respiratory muscle strength and pulmonary function in judokas, especially those with pre-existing EIB, thereby prolonging spontaneous recovery time.
Collapse
Affiliation(s)
- Özgür Bostancı
- Faculty of Sport Sciences, University of Ondokuz Mayıs, Samsun, Türkiye ; and
| | - Emre Karaduman
- Faculty of Sport Sciences, University of Ondokuz Mayıs, Samsun, Türkiye ; and
| | - Ali Kerim Yılmaz
- Faculty of Sport Sciences, University of Ondokuz Mayıs, Samsun, Türkiye ; and
| | - Menderes Kabadayı
- Faculty of Sport Sciences, University of Ondokuz Mayıs, Samsun, Türkiye ; and
| | - Sait Bilgiç
- Faculty of Medicine, University of Ondokuz Mayıs, Samsun, Türkiye
| |
Collapse
|
16
|
Abstract
Coronavirus disease 2019 (COVID-19) remains a health problem worldwide. The present study aimed to investigate the effect of blood pressure (BP) on the circadian pattern and prevalence of new-onset non-dipper hypertension in the post-COVID period in patients with known hypertension. This prospective single-center study included 722 patients hospitalized for COVID-19 infection. Ambulatory BP (ABP) data were collected during their initial hospitalization. The ABP data were reassessed 1 month after the patients were discharged. The results were compared with a healthy control group with known hypertension but without COVID-19 infection. After exclusion criteria were applied, the study included 187 patients with COVID-19 and 136 healthy hypertensive controls. Post-COVID ABP showed that patients with COVID-19 had significantly higher mean 24-h systolic and diastolic BP, mean nighttime systolic and diastolic BP, and mean daytime diastolic BP than the control group. In addition, new-onset non-dipper hypertension was significantly higher in patients with COVID-19. This study demonstrated for the first time that the circadian pattern is disturbed and a non-dipper pattern develops in individuals with known hypertension during the post-COVID period.
Collapse
Affiliation(s)
- Fatih Sivri
- Aydin Nazilli State Hospital, Nazilli, Turkey
| | - Ismail Türköz
- Department of Infectious Diseases, Dortyol State Hospital, Hatay, Turkey
| | - Mehtap Şencan
- Department of Infectious Diseases, Dortyol State Hospital, Hatay, Turkey
| | - Yahya Kemal İçen
- Department of Cardiology, Adana Health Practice and Research, Adana, Turkey
| | - Fatih Aksoy
- Department of Cardiology, Süleyman Demirel University, Isparta, Turkey
| | - Banu Öztürk Ceyhan
- Department Of Endocrine Diseases, Adnan Menderes University, Aydın, Turkey
| |
Collapse
|
17
|
Aliberti A, Gasparro R, Mignogna M, Canfora F, Spagnuolo G, Sammartino G, Coppola N. Unveiling the Oral Lesions, Dysgeusia and Osteonecrosis Related to COVID-19: A Comprehensive Systematic Review. J Clin Med 2025; 14:1267. [PMID: 40004799 PMCID: PMC11856591 DOI: 10.3390/jcm14041267] [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: 02/01/2025] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The oral cavity has garnered increasing attention as a site for viral infection and related pathological manifestations in coronavirus disease-19. This article aims to provide a comprehensive overview of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2)-related oral manifestations, including taste disturbances, oral lesions and osteonecrosis. Methods: A search was conducted up to September 2024 according to PRISMA (Preferred Reporting Items for Systematic Reviews) guidelines using the databases PubMed and Scopus. All the observational, case-series, case-report and cross-sectional studies written in English on oral manifestations related to COVID-19 disease and long-COVID disease were included. All other types of studies and studies based on oral manifestation after COVID-19 vaccination and oral impairment due to lockdown were excluded. The risk of bias of included studies was assessed using the Joanna Briggs Appraisal checklist. Results: A total of 104 articles including 23 case-report, 15 case-series, 8 case-control, 18 cohort and 40 cross-sectional studies were selected. The results showed that patients with COVID-19 were found to have a significantly higher prevalence of xerostomia (45-74%) and dysgeusia (32-59%) compared to non-infected individuals. Regarding oral mucosal lesions, ulcers, candidiasis and herpes simplex infections were frequently observed. As for osteonecrosis, a significant number of patients with COVID-19-associated rhinomaxillary mucormycosis presented with maxillary osteonecrosis due to fungal infection, primarily mucormycosis. The methodological quality of most of the studies was moderate/high. Conclusions: COVID-19 has been associated with a range of oral manifestations. The complex interplay of viral infection, immune response, medication use and stress likely contributes to these oral complications. Early recognition and management of these oral manifestations are crucial for improving patient outcomes and developing targeted preventive and therapeutic strategies for COVID-19-related oral health issues.
Collapse
Affiliation(s)
- Angelo Aliberti
- Department of Neurosciences, Reproductive Sciences and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.A.); (R.G.); (M.M.); (F.C.); (G.S.); (N.C.)
| | - Roberta Gasparro
- Department of Neurosciences, Reproductive Sciences and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.A.); (R.G.); (M.M.); (F.C.); (G.S.); (N.C.)
| | - Martina Mignogna
- Department of Neurosciences, Reproductive Sciences and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.A.); (R.G.); (M.M.); (F.C.); (G.S.); (N.C.)
| | - Federica Canfora
- Department of Neurosciences, Reproductive Sciences and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.A.); (R.G.); (M.M.); (F.C.); (G.S.); (N.C.)
| | - Gianrico Spagnuolo
- Department of Neurosciences, Reproductive Sciences and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.A.); (R.G.); (M.M.); (F.C.); (G.S.); (N.C.)
- Therapeutic Dentistry Department, Institute for Dentistry, Sechenov University, Moscow 119991, Russia
| | - Gilberto Sammartino
- Department of Neurosciences, Reproductive Sciences and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.A.); (R.G.); (M.M.); (F.C.); (G.S.); (N.C.)
| | - Noemi Coppola
- Department of Neurosciences, Reproductive Sciences and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.A.); (R.G.); (M.M.); (F.C.); (G.S.); (N.C.)
| |
Collapse
|
18
|
Voros C, Mavrogianni D, Minaoglou A, Papahliou AM, Topalis V, Varthaliti A, Mathiopoulos D, Kondili P, Darlas M, Pantou A, Sina S, Athanasiou A, Athanasiou D, Loutradis D, Daskalakis G. Unveiling the Impact of COVID-19 on Ovarian Function and Premature Ovarian Insufficiency: A Systematic Review. Biomedicines 2025; 13:407. [PMID: 40002820 PMCID: PMC11853103 DOI: 10.3390/biomedicines13020407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Premature ovarian insufficiency (POI) is a disorder that affects women under the age of 40. It is characterized by decreased ovarian function, elevated gonadotropin levels, and decreased estradiol. SARS-CoV-2 disrupts ovarian function largely through oxidative stress, inflammation, and immunological dysregulation, which are enhanced by its entrance into ovarian tissues via ACE2 receptors. The purpose of this comprehensive review was to investigate the molecular pathways that link SARS-CoV-2 infection to POI and analyze their consequences for ovarian reserve and fertility. Methods: We searched databases such as PubMed, Scopus, EMBASE, and Google Scholar for papers published between 2020 and 2024. Eligible studies investigated the effects of SARS-CoV-2 on ovarian function, including the hormonal indicators anti-Müllerian hormone (AMH) and follicle-stimulating hormone (FSH), oocyte quality, and ovarian reserve. The data were compiled into a complete examination of molecules and clinical findings. Increased inflammatory indicators, such as interleukin-6 and NLRP3 inflammasome activation, impaired ovarian homeostasis. Anti-SARS-CoV-2 antibodies in follicular fluid could have impaired oocyte quality. Observational studies showed transitory decreases in AMH and changed FSH levels following infection, with variable effects on antral follicle count and IVF results. Changes in lipid profiles and VEGF expression emphasized the virus's influence on ovarian angiogenesis and the ovarian microenvironment. Conclusions: SARS-CoV-2 infection impairs ovarian function by causing oxidative stress, inflammation, and hormonal disruption, thereby increasing the incidence of POI. While most alterations are temporary, the long-term reproductive consequences remain unknown. Continuous monitoring and specific treatments are required to reduce the reproductive risks associated with COVID-19.
Collapse
Affiliation(s)
- Charalampos Voros
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.M.); (A.-M.P.); (A.V.); (P.K.); (M.D.); (A.P.); (S.S.); (G.D.)
| | - Despoina Mavrogianni
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.M.); (A.-M.P.); (A.V.); (P.K.); (M.D.); (A.P.); (S.S.); (G.D.)
| | - Aspasia Minaoglou
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.M.); (A.-M.P.); (A.V.); (P.K.); (M.D.); (A.P.); (S.S.); (G.D.)
| | - Anthi-Maria Papahliou
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.M.); (A.-M.P.); (A.V.); (P.K.); (M.D.); (A.P.); (S.S.); (G.D.)
| | - Vasileios Topalis
- Department of Internal Medicine, Hospital of Thun, 3600 Thun, Switzerland;
| | - Antonia Varthaliti
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.M.); (A.-M.P.); (A.V.); (P.K.); (M.D.); (A.P.); (S.S.); (G.D.)
| | - Dimitris Mathiopoulos
- Rea Maternity Hospital S.A., Avenue Siggrou 383 & Pentelis 17, P. Faliro, 17564 Athens, Greece;
| | - Panagiota Kondili
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.M.); (A.-M.P.); (A.V.); (P.K.); (M.D.); (A.P.); (S.S.); (G.D.)
| | - Menelaos Darlas
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.M.); (A.-M.P.); (A.V.); (P.K.); (M.D.); (A.P.); (S.S.); (G.D.)
| | - Agni Pantou
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.M.); (A.-M.P.); (A.V.); (P.K.); (M.D.); (A.P.); (S.S.); (G.D.)
| | - Sophia Sina
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.M.); (A.-M.P.); (A.V.); (P.K.); (M.D.); (A.P.); (S.S.); (G.D.)
| | - Antonia Athanasiou
- IVF Athens Reproduction Center V. Athanasiou, 15123 Maroussi, Greece; (A.A.); (D.A.)
| | - Diamantis Athanasiou
- IVF Athens Reproduction Center V. Athanasiou, 15123 Maroussi, Greece; (A.A.); (D.A.)
| | - Dimitrios Loutradis
- Fertility Institute-Assisted Reproduction Unit, Paster 15, 11528 Athens, Greece;
- Athens Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Georgios Daskalakis
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.M.); (A.-M.P.); (A.V.); (P.K.); (M.D.); (A.P.); (S.S.); (G.D.)
| |
Collapse
|
19
|
Yang G, Zhu L, Wang Y, Yu Y, Liu X, Xia J, Yang Y, Wang H, Feng B. Antihypertensive effect of sinapine extracted from rapeseed meal in 2K1C hypertensive rats. Sci Rep 2025; 15:4133. [PMID: 39900955 PMCID: PMC11790856 DOI: 10.1038/s41598-025-88926-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 01/31/2025] [Indexed: 02/05/2025] Open
Abstract
To extract sinapine from rapeseed meal and investigate its antihypertensive function and mechanism. Blood pressure was measured before and after sinapine administration to evaluate sinapine's immediate antihypertensive function. Twokidney, oneclip (2K1C) hypertensive rats were given sinapine for four weeks, with weekly blood pressure monitoring. The renin angiotensin aldosterone system (RAAS), including the levels of renin, angiotensin I (Ang I), angiotensin II (Ang II), aldosterone (ALD), angiotensin-converting enzyme (ACE) and other molecules related to blood pressure, such as NO, prostacyclin (PGI2), endothelin-1 (ET1), and thromboxane A2 (TXA2), were measured in rat blood. The impact of sinapine on vascular endothelial cell (A10) calcium and potassium channels was assessed using the patch-clamp technique. One-time or long-term administration of sinapine significantly reduced the rats' systolic blood pressure (SBP), diastolic pressure (DBP), and mean blood pressure (MBP). Sinapine also decreased the levels of Ang II and ALD. Furthermore, sinapine effectively inhibited ACE activation, increased NO levels, and blocked L-type calcium channels. Sinapine has an antihypertensive function and achieves this process through multiple targets.
Collapse
Affiliation(s)
- Gaoyuan Yang
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Lin Zhu
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Yu Wang
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Yang Yu
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Xiaoguang Liu
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Jingbo Xia
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Yunjie Yang
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Huiguo Wang
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China.
| | - Baomin Feng
- College of Life and Health, Dalian University, Liaoning, Dalian, 116622, China.
| |
Collapse
|
20
|
Abodunrin OR, Olagunju MT, Huang X, Wang J, Hu Z, Shen C. Regional risk factors associated with adverse outcomes of COVID-19 infection among the older adult: A systematic review and meta-analysis. J Infect Public Health 2025; 18:102632. [PMID: 39754850 DOI: 10.1016/j.jiph.2024.102632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025] Open
Abstract
The rapid global spread of Coronavirus Disease 2019 (COVID-19) has resulted in millions of infections and deaths, particularly impacting older adults. This study systematically analyzes risk factors reported in different geographical regions such as Asia and Europe that are associated with adverse outcomes in older adults with COVID-19. Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we searched five databases up to December 2023 and conducted meta-analyses of odds ratios for 27 risk factors reported in at least two studies using R software (version 4.3.2). Our meta-analysis identified 19 risk factors linked to adverse outcomes, with many of them common across regions, particularly in Asia and Europe. Key factors include old age (above 65 years), male gender, symptoms such as fever and dyspnea, and comorbidities like dementia, chronic obstructive pulmonary disease (COPD), chronic heart disease, hypertension, chronic kidney disease, and malnutrition. Laboratory biomarkers such as low oxygen saturation, thrombocytopenia, and elevated D-dimer were also associated with adverse outcomes. COVID-19 patients in Asia and Europe who are older adults, male, or have specific symptoms combined with underlying health conditions are at an increased risk of progressing to severe illness or mortality.
Collapse
Affiliation(s)
- Olunike Rebecca Abodunrin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Mobolaji Timothy Olagunju
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xinyi Huang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jianming Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhibin Hu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Chong Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
21
|
Shaik KM, Kumar D, Srikanth P, Nandi S. SARS-CoV-2: A synergy to the Alzheimer's disease. J Neurovirol 2025; 31:16-23. [PMID: 39998800 DOI: 10.1007/s13365-025-01247-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
COVID-19 was a nightmare in humankind's history that challenged our advanced medical technology. All credit goes to the researchers who played a crucial role in curbing COVID-19 and proved our medical technology supremacy. However, COVID-19 has left some mysterious scars on human well-being. It is believed that COVID-19 has a significant negative impact on various cardiovascular (CVS) and central nervous system (CNS) diseases, especially in the case of CNS diseases like Alzheimer's. Surprisingly, COVID-19 affects the respiratory system, whereas Alzheimer's disease (AD) alters brain function. To explain this phenomenon, several hypotheses were proposed, but the mechanism needs to be clearly understood. Another critical thing to be concerned about is that COVID-19 will worsen pre-existing conditions and lead to the onset of AD. In the race to curb COVID-19, the invention of vaccines was speeded up, and it is necessary to fight against COVID-19. However, postvaccination follow-up is mandatory when an individual is a victim of AD. In this review article, we compiled the various dreadful effects of the COVID-19 virus on AD, the Post effects of the virus on AD, and the effect of the COVID-19 vaccination on AD. This article provides a new direction for research concerning COVID-19 and AD.
Collapse
Affiliation(s)
- Khaja Moinuddin Shaik
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Deepak Kumar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Pirangi Srikanth
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Sukhendu Nandi
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, 160 062, Punjab, India.
| |
Collapse
|
22
|
Zima K, Bogucka A, Wojtas M, Zabielska-Kaczorowska M. Immunological Effects of Electronic Cigarette Use: A Review of Current Evidence. Clin Rev Allergy Immunol 2025; 68:9. [PMID: 39891861 DOI: 10.1007/s12016-025-09026-5] [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] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
Electronic cigarettes (EC) have emerged as a popular alternative to traditional tobacco products, but their impact on immune function has raised significant health concerns. This review explores the immunological effects of EC exposure, focusing on innate and adaptive immune responses. Electronic cigarette aerosol (ECA) induces widespread inflammation. These changes compromise immune cell function, impairing neutrophil chemotaxis, phagocytosis, and oxidative burst while increasing macrophage and dendritic cell recruitment and activation. ECA also disrupts epithelial barriers, increasing susceptibility to bacterial and viral infections. Studies show enhanced biofilm formation in bacteria such as Staphylococcus aureus and Streptococcus pneumoniae and impaired antiviral responses against pathogens like influenza A and SARS-CoV-2. Additionally, EC exposure modulates adaptive immunity, affecting T and B cell function and increasing systemic inflammatory markers. The long-term consequences of these immunological disruptions include heightened risks for chronic inflammatory diseases, respiratory infections, and potentially autoimmune conditions. The widespread adoption of EC, particularly among younger users, poses a growing public health challenge. As the popularity of vaping continues to rise, these immunological disruptions could result in increased healthcare burdens in the future, with higher rates of infections, chronic inflammatory diseases, and immune system-related disorders among those who begin using e-cigarettes at a young age. Understanding the full scope of EC-related health risks is essential for informing public health policies and protecting future generations from the potential long-term effects of vaping.
Collapse
Affiliation(s)
- Katarzyna Zima
- Department of Physiology, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland.
| | - Aleksandra Bogucka
- Department of Physiology, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Miłosz Wojtas
- Department of Physiology, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | | |
Collapse
|
23
|
Pogreba Brown K, Austhof E, McFadden CM, Scranton C, Sun X, Vujkovic-Cviji I, Rodriguez D, Falk L, Heslin KM, Arani G, Obergh V, Bessey K, Cooper K. Determining the incidence, risk factors and biological drivers of irritable bowel syndrome (IBS) as part of the constellation of postacute sequelae of SARS-CoV-2 infection (PASC) outcomes in the Arizona CoVHORT-GI: a longitudinal cohort study. BMJ Open 2025; 15:e095093. [PMID: 39890144 PMCID: PMC11784208 DOI: 10.1136/bmjopen-2024-095093] [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: 10/15/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025] Open
Abstract
INTRODUCTION Postacute sequelae of SARS-CoV-2 infection (PASC) are extensive. Also known as long COVID, primary outcomes reported are neurologic, cardiac and respiratory in nature. However, several studies have also reported an increase in gastrointestinal (GI) symptoms and syndromes following COVID-19. This study of PASC will include extensive analyses of GI symptoms, determine if people with pre-existing irritable bowel syndrome (IBS) are at higher risk of developing PASC generally or PASC-GI, and which biomarkers are impacted and to what degree. This R01 study is being funded by the National Institute of Diabetes and Digestive and Kidney Diseases (1R01DK135483-01) from 2023 to 2028. METHODS AND ANALYSES This study combines a longitudinal epidemiologic cohort study and in-depth, novel biologic analyses. In collaboration with a pre-existing study, the Arizona CoVID-19 Cohort (CoVHORT)-GI will recruit participants based on the history of COVID infection(s), new or ongoing GI symptoms 3-6 months postinfection, and pre-existing or incident IBS diagnosis to represent five study groups for comparison and analyses. A subset (n=1000) of those recruited will submit both stool and blood samples. Both samples will undergo a novel method to quantitate humoral and mucosal immune responses to host-derived faecal communities in conjunction with magnetic bead-based separation and high-depth shotgun microbial sequencing. Stool samples will also undergo traditional microbiome analyses (diversity and abundance) and faecal calprotectin assays. Additional serum analyses will aim to determine if a proteomics-based signature exists that differentiates a unique biomarker compositional signature discriminating PASC-GI versus no PASC. All laboratory data will be linked with in-depth epidemiologic data on demographics, symptoms and chronic conditions. ETHICS AND DISSEMINATION This study involves human participants and was approved by the University of Arizona Institutional Review Board (IRB (#00002332) and has been deemed minimal risk. Participants gave informed consent to participate in the study before taking part. All publications from the study will be shared back to participants along with alternative lay summaries and webinars to communicate key findings. The data management plan has been published and is publicly available online, including protocols for data requests.
Collapse
Affiliation(s)
- Kristen Pogreba Brown
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Erika Austhof
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Caitlyn M McFadden
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Caroline Scranton
- School of Animal and Comparative Biomedical Sciences, The University of Arizona College of Agriculture and Life Sciences, Tucson, Arizona, USA
| | - Xiaoxiao Sun
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Ivan Vujkovic-Cviji
- Department of Biomedical Sciences; Research Division of Immunology, Department of Medicine, Karsh Division of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dominic Rodriguez
- School of Animal and Comparative Biomedical Sciences, The University of Arizona College of Agriculture and Life Sciences, Tucson, Arizona, USA
| | - Laura Falk
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Kelly M Heslin
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Gayatri Arani
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona, Arizona Health Sciences Center, Tucson, Arizona, USA
| | - Victoria Obergh
- School of Animal and Comparative Biomedical Sciences, The University of Arizona College of Agriculture and Life Sciences, Tucson, Arizona, USA
| | - Kate Bessey
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Kerry Cooper
- School of Animal and Comparative Biomedical Sciences, The University of Arizona College of Agriculture and Life Sciences, Tucson, Arizona, USA
| |
Collapse
|
24
|
de Souza T, Rosa AS, Constantino-Teles P, Ferreira VNS, Archanjo BS, Soares CAG, Picciani PHS, Allão Cassaro RA, Miranda MD, Poneti G. Silver Nanoparticles-Functionalized Textile against SARS-CoV-2: Antiviral Activity of the Capping Oleylamine Molecule. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5710-5718. [PMID: 39807796 PMCID: PMC11788990 DOI: 10.1021/acsami.4c15289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
COVID-19 disease, triggered by SARS-CoV-2 virus infection, has led to more than 7.0 million deaths worldwide, with a significant fraction of recovered infected people reporting postviral symptoms. Smart surfaces functionalized with nanoparticles are a powerful tool to inactivate the virus and prevent the further spreading of the disease. Literature reports usually focus on the role of nanomaterial composition and size dispersion in evaluating their efficacy against SARS-CoV-2. Here, the anti-SARS-CoV-2 activity of oleylamine (OAm) used as a capping agent of silver nanoparticles is quantified for the first time. Spherical hydrophobic nanoparticles with 8 ± 2 nm diameter were prepared and characterized by Fourier transform infrared, dynamic light scattering, and transmission electron microscopy techniques. Biological assays showed that microgram amounts of nanoparticles, deposited on nonwoven textile obtained from surgical masks, efficiently inactivated up to 99.6(2)% of the virus with just 2 min of exposure. The virucidal activity of the corresponding amount of free OAm has been determined as well, reaching up to 67(1)% of activity for an exposure time of 10 min. Inductively coupled plasma optical emission spectrometry results pointed out a low leaching out of the nanoparticles in contact with water or culture medium. All in all, these results propose the capping molecules as an important chemical variable to be taken into account in the design of fast, efficient, and long-lasting anti-SARS-CoV-2 coatings.
Collapse
Affiliation(s)
| | - Alice S. Rosa
- Laboratory
of Morphology and Virus Morphogenesis, Oswaldo
Cruz Institute, Fiocruz,
Avenida Brasil, Rio de Janeiro 21041-250, Brazil
- Programa
de pós-graduação em Biologia Celular e Molecular,
Instituto Oswaldo Cruz, Fundação
Oswaldo Cruz, Rio de
Janeiro 21041-250, Brazil
| | - Pamella Constantino-Teles
- Laboratory
of Morphology and Virus Morphogenesis, Oswaldo
Cruz Institute, Fiocruz,
Avenida Brasil, Rio de Janeiro 21041-250, Brazil
- Programa
de pós-graduação em Biologia Celular e Molecular,
Instituto Oswaldo Cruz, Fundação
Oswaldo Cruz, Rio de
Janeiro 21041-250, Brazil
| | - Vivian Neuza S. Ferreira
- Laboratory
of Morphology and Virus Morphogenesis, Oswaldo
Cruz Institute, Fiocruz,
Avenida Brasil, Rio de Janeiro 21041-250, Brazil
| | - Braulio S. Archanjo
- Materials
Metrology Division, National Institute of
Metrology, Quality, and Technology, Duque de Caxias, Rio de Janeiro 25250-020, Brazil
| | - Carlos A. G. Soares
- Departamento
de Genética, Universidade Federal
do Rio de Janeiro, Rio de
Janeiro 21941-617, Brazil
| | - Paulo H. S. Picciani
- Instituto
de Macromoléculas Professora Eloisa Mano, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Rafael A. Allão Cassaro
- Instituto
de Química, Universidade Federal
do Rio de Janeiro, Rio de
Janeiro 21941-909, Brazil
| | - Milene Dias Miranda
- Laboratory
of Morphology and Virus Morphogenesis, Oswaldo
Cruz Institute, Fiocruz,
Avenida Brasil, Rio de Janeiro 21041-250, Brazil
- Programa
de pós-graduação em Biologia Celular e Molecular,
Instituto Oswaldo Cruz, Fundação
Oswaldo Cruz, Rio de
Janeiro 21041-250, Brazil
| | - Giordano Poneti
- Instituto
de Química, Universidade Federal
do Rio de Janeiro, Rio de
Janeiro 21941-909, Brazil
- Dipartimento
di Scienze Ecologiche e Biologiche, Università
degli Studi della Tuscia, Largo dell’Università, Viterbo 01100, Italy
| |
Collapse
|
25
|
Huang J, Fan Y, Wang Y, Liu J. The effects of enhanced external counter-pulsation on post-acute sequelae of COVID-19: A narrative review. Open Med (Wars) 2025; 20:20241067. [PMID: 39802655 PMCID: PMC11716443 DOI: 10.1515/med-2024-1067] [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: 05/17/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 01/16/2025] Open
Abstract
Some of the millions of patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have developed new sequelae after recovering from the initial disease, termed post-acute sequelae of coronavirus disease 2019 (PASC). One symptom is anxiety, which is likely due to three etiologies: brain structural changes, neuroendocrine disruption, and neurotransmitter alterations. This review provides an overview of the current literature on the pathophysiological pathways linking coronavirus disease 2019 to anxiety, as well as the possible mechanisms of action in which an increasingly scrutinized treatment method, enhanced external counter-pulsation (EECP), is able to alleviate anxiety. SARS-CoV-2 triggers increased inflammatory cytokine production, as well as oxidative stress; these processes contribute to the aforementioned three etiologies. The potential treatment approach of EECP, involving sequenced inflation and deflation of specifically-placed airbags, has become of increasing interest, as it has been found to alleviate PASC-associated anxiety by improving patient cardiovascular function. These functional improvements were achieved by EECP stimulating anti-inflammatory and pro-angiogenic processes, as well as improving endothelial cell function and coronary blood flow, partially via counteracting against the negative effects of SARS-CoV-2 infection on the renin-angiotensin-aldosterone system. Therefore, EECP could promote both psychosomatic and cardiac rehabilitation. Further research, though, is still needed to fully determine its benefits and mechanism of action.
Collapse
Affiliation(s)
- Jiecheng Huang
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
| | - Yuxuan Fan
- Department of Rehabilitation, Tongji Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai, China
| | - Yongshun Wang
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
- Department of Cardiology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Jingjin Liu
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
- Department of Cardiology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| |
Collapse
|
26
|
Sayers EW, Cavanaugh M, Frisse L, Pruitt KD, Schneider VA, Underwood B, Yankie L, Karsch-Mizrachi I. GenBank 2025 update. Nucleic Acids Res 2025; 53:D56-D61. [PMID: 39558184 PMCID: PMC11701615 DOI: 10.1093/nar/gkae1114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/20/2024] Open
Abstract
GenBank® (https://www.ncbi.nlm.nih.gov/genbank/) is a comprehensive, public data repository that contains 34 trillion base pairs from over 4.7 billion nucleotide sequences for 581 000 formally described species. Daily data exchange with the European Nucleotide Archive and the DNA Data Bank of Japan ensures worldwide coverage. We summarize the content of the database in 2025 and recent updates such as accelerated processing of influenza sequences and the ability to upload feature tables to Submission Portal for messenger RNA sequences. We provide an overview of the web, application programming and command-line interfaces that allow users to access GenBank data. We also discuss the importance of creating BioProject and BioSample records during submissions, particularly for viruses and metagenomes. Finally, we summarize educational materials and recent community outreach efforts.
Collapse
Affiliation(s)
- Eric W Sayers
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Building 38A, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Mark Cavanaugh
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Building 38A, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Linda Frisse
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Building 38A, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Kim D Pruitt
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Building 38A, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Valerie A Schneider
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Building 38A, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Beverly A Underwood
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Building 38A, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Linda Yankie
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Building 38A, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Ilene Karsch-Mizrachi
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Building 38A, 8600 Rockville Pike, Bethesda, MD 20894, USA
| |
Collapse
|
27
|
Almalki SK, Azzam AM, Alhammad SA, Alabdulwahab S, Alshamrani AA, Alotaibi AN. Outcomes of a Structured Olfactory and Gustatory Rehabilitation Program in Children with Post-COVID-19 Smell and Taste Disturbances. J Clin Med 2025; 14:272. [PMID: 39797354 PMCID: PMC11722520 DOI: 10.3390/jcm14010272] [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: 12/03/2024] [Revised: 12/21/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is closely related to SARS-CoV and uses angiotensin-converting enzyme 2 as its cellular receptor. In early 2020, reports emerged linking CoV disease 2019 (COVID-19) to olfactory and gustatory disturbances. These disturbances could be attributed to virus-induced damage to olfactory neurons or immune responses, thereby affecting sensory functions. This randomized controlled trial aimed to evaluate the effectiveness of a structured orofacial rehabilitation program in improving smell (olfaction) and taste (gustation) sensations in children post-COVID-19. Methods: Forty children recovering from COVID-19 in government hospitals in Saudi Arabia were included and randomly assigned to the control group or the experimental group. The orofacial program included (a) facilitation of olfactory function using the 40-item modified Arabic version of the University of Pennsylvania Smell Identification Test (UPSIT); (b) assessment of gustatory function using taste strips with four varying concentrations; and (c) orofacial myofunctional therapy. The intervention was applied three times a week and lasted for 3 months. Results: The experimental group showed a significantly greater improvement in UPSIT scores (median change of 24.1%) than the control group (14.7%; p = 0.010). However, no significant difference was found in the taste strip test scores among the groups or between male and female participants. Conclusions: This study suggests that a structured orofacial rehabilitation program could enhance olfactory and gustatory functions in children recovering from COVID-19.
Collapse
Affiliation(s)
- Smai Khalid Almalki
- King Abdulaziz Specialist Hospital, Taif Health Cluster, Taif 26521, Saudi Arabia;
| | - Ahmed Mohamed Azzam
- Department of Physiotherapy for Developmental Disturbance and Pediatric Surgery, Faculty of Physical Therapy, Cairo University, Giza 12613, Egypt;
| | - Saad A. Alhammad
- Department of Rehabilitation Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia;
| | - Sami Alabdulwahab
- Department of Rehabilitation Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia;
| | - Ahmed Ali Alshamrani
- King Abdulaziz Specialist Hospital, Taif Health Cluster, Taif 26521, Saudi Arabia;
| | | |
Collapse
|
28
|
Al Ghobain M, Farahat F, Zeitouni M, Alsowayan W, Al-Awfi S, AlBarrak A, Al-Basheri S, Alhabeeb F, Alhamad EH. The Saudi thoracic society guidelines for vaccinations in adult patients with chronic respiratory diseases. Ann Thorac Med 2025; 20:36-48. [PMID: 39926401 PMCID: PMC11804953 DOI: 10.4103/atm.atm_202_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 02/11/2025] Open
Abstract
Adult patients with chronic respiratory diseases (CRDs) are considered high risk group who are more likely to experience worse clinical outcomes if they acquire viral or bacterial infections. Vaccination is the best preventive tool to reduce the risk of infection and disease occurrence and to reduce the level of severity of complications associated with the various vaccine-preventable infections. These guidelines were developed by the Saudi Thoracic Society task force to emphasize the critical importance of improving the vaccine coverage rates in adult patients with CRD. They are intended to serve as a reference for healthcare practitioners managing CRD patients. The guidelines aimed to review the current knowledge related to vaccination efficacy in adult patients with CRD, based on the recent evidence and recommendations. Integrating the administration of the recommended vaccines in routine healthcare, such as during outpatient visits or before hospital discharge, is crucial for improving the vaccination rates in high-risk patients. The key strategies to address this public health priority include simplifying vaccination guidelines to enhance their accessibility and implementation by healthcare providers, increasing awareness in both the patients and healthcare providers that vaccines are not only intended for children. Additional strategies include maintaining continuous surveillance and advance research to discover novel vaccines. This approach aims to expand the range of preventable diseases and improve overall health and well-being. Vaccine hesitancy remains a significant challenge that necessitates a clear understanding of the community concerns. Providing appropriate education and communication, as well as addressing these concerns, are the crucial steps toward improving vaccine acceptance and uptake. By implementing these guidelines and multifaceted strategies, healthcare systems can optimize vaccine coverage and protection for patients with CRD, reduce the burden of vaccine-preventable complications, and improve the clinical outcomes in this vulnerable population.
Collapse
Affiliation(s)
- Mohammed Al Ghobain
- Professor of Pulmonary Medicine, King Abdulaziz Medical City, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Fayssal Farahat
- Director, Community and Public Health, King Abdulaziz Medical City, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mohammed Zeitouni
- Consultant Pulmonologist, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Waleed Alsowayan
- Consultant Pulmonologist, Security Forces Hospital, Riyadh, Saudi Arabia
| | - Sultan Al-Awfi
- Consultant Infectious Diseases, Security Forces Hospital, Riyadh, Saudi Arabia
| | - Ali AlBarrak
- Consultant Infectious Diseases, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Shareefah Al-Basheri
- Consultant Pulmonologist, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Fatmah Alhabeeb
- Consultant Pulmonologist, King Khalid University Hospital, Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Esam H. Alhamad
- Professor of Pulmonary Medicine, Department of Medicine, Division of Pulmonary Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
29
|
Aleissa M, Drelichman E, Bhullar J. Ischemic Colitis and Small Bowel Ischemia in a Vaccinated Patient with Mild COVID-19 Infection: A Case Report. Case Rep Gastroenterol 2025; 19:335-339. [PMID: 40351853 PMCID: PMC12064152 DOI: 10.1159/000545495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/17/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction COVID-19 infection can cause bowel ischemia, with an incidence ranging from 0.22% to 10.5. The COVID-19 vaccine reduces respiratory symptoms and the need for cardiopulmonary support. However, its effects on other manifestations, such as bowel ischemia, have not been extensively studied. Despite having mild respiratory symptoms and receiving three doses of the COVID-19 vaccine, our patient developed ischemic colitis after her first infection and small bowel ischemia following her second infection. Case Presentation An 86-year-old woman presented to the emergency department (ED) with abdominal pain after a mild COVID-19 infection. She was admitted with ischemic colitis, but conservative treatment failed. In the operating room, her entire colon was found to be ischemic, necessitating a total colectomy with end ileostomy. Nine months later, she returned to the ED with mild respiratory symptoms and severe right upper quadrant pain. Imaging revealed pneumoperitoneum and a mid-abdominal abscess. An emergency laparotomy revealed small bowel perforation, with final histology confirming ischemia. The patient had received three doses of the COVID-19 vaccine and was on therapeutic anticoagulation for a history of pulmonary embolism. Conclusion COVID-19 may increase the risk of bowel and colon ischemia even after vaccination. Patients presenting to the ED with severe abdominal pain and a recent COVID-19 infection should be carefully evaluated for ischemic events.
Collapse
Affiliation(s)
- Maryam Aleissa
- Department of Surgery, Henry Ford Providence Hospital, Collage of Humen Medicine, Michigan State University, Southfield, MI, USA
- College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ernesto Drelichman
- Department of Surgery, Henry Ford Providence Hospital, Collage of Humen Medicine, Michigan State University, Southfield, MI, USA
| | - Jasneet Bhullar
- Department of Surgery, Henry Ford Providence Hospital, Collage of Humen Medicine, Michigan State University, Southfield, MI, USA
| |
Collapse
|
30
|
Majumder MH, Sazzad S, Hasin R, Brishti TJ, Tabassum FN, Ahamed T, Masud AA, Akter F. The Impact of Low Serum Magnesium Levels on COVID-19 Severity and Potential Therapeutic Benefits of Magnesium Supplementation: A Systematic Review. Cureus 2025; 17:e77118. [PMID: 39925614 PMCID: PMC11803333 DOI: 10.7759/cureus.77118] [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: 01/07/2025] [Indexed: 02/11/2025] Open
Abstract
In this review, our objective was to analyze the association between serum magnesium (Mg) levels, Mg supplementation, and coronavirus disease 2019 (COVID-19) outcomes. This systematic review followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, searching major databases until February 2023. Twenty-six studies (11,363 patients) were included: 22 examining serum Mg levels (8474 patients) and four investigating Mg supplementations (2889 patients). Most studies indicated an association between lower serum Mg levels and increased COVID-19 severity, including higher mortality rates and prolonged recovery periods. Critical patients demonstrated significantly lower Mg levels compared to moderate/severe cases. However, some studies reported conflicting findings, with hypermagnesemia also associated with poor outcomes in specific patient populations. Regarding supplementation, higher dietary Mg intake correlated with shorter hospitalization duration and faster recovery. Mg supplementation exceeding 450 mg showed potential benefits, including increased antibody titers in pregnant women and reduced oxygen support requirements in elderly patients when combined with vitamins D and B12. While evidence suggests a potential relationship between Mg status and COVID-19 outcomes, findings are heterogeneous. Further investigation through well-designed clinical trials is required to gain deeper insights into the role of Mg in COVID-19 pathophysiology and the therapeutic potential of Mg supplementation.
Collapse
Affiliation(s)
| | - Sadman Sazzad
- Health Informatics, Indiana University Indianapolis, Indianapolis, USA
| | - Rabeya Hasin
- Medicine, Dhaka Medical College and Hospital, Dhaka, BGD
| | | | | | - Tanvir Ahamed
- Medicine, Army Medical College Cumilla, Cumilla, BGD
| | - Abdullah A Masud
- Internal Medicine, Saint Louis University School of Medicine, St. Louis, USA
| | - Fahima Akter
- Internal Medicine, Indiana University Health Ball Memorial Hospital, Muncie, USA
| |
Collapse
|
31
|
Wen R, Li J, Chen F, Liu J, Xu P, Li M, Li J, Tan L, Liu C. Effect of the renin-angiotensin-aldosterone system inhibitors on time to nucleic acid negative conversion in hypertensive patients with SARS-CoV-2 omicron infection: a propensity score matching study. Hypertens Res 2025; 48:273-283. [PMID: 39506050 DOI: 10.1038/s41440-024-01953-8] [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: 03/08/2024] [Revised: 09/17/2024] [Accepted: 10/05/2024] [Indexed: 11/08/2024]
Abstract
We examined the relationship between RAAS inhibitor use and Omicron infection in mildly symptomatic patients. This retrospective case-control observational study included 50,121 patients with mild Omicron infection from the largest "Fangcang" shelter hospital in Shanghai between April 9, 2022, and May 21, 2022. Using 1:1 propensity score matching (PSM), we classified 4394 COVID-19 patients into hypertension and non-hypertension groups, and 406 hypertensive patients into RAAS inhibitor and non-RAAS inhibitor groups. The risk of initial symptoms of infection, cumulative negative conversion rates, time to nucleic-acid negative conversion, and viral loads were compared. In the hypertension group, the median number of days for nucleic-acid negative conversion was 7.0 (IQR, 5.0-9.0), which was greater than non-hypertensive group (median (IQR) 6.0 (4.0-8.0), P < 0.001, Cohen's d = 0.29); the mean and minimum cycle threshold values (CT-values) were significantly lower (P < 0.001, Cohen's d = 0.23). In the RAAS inhibitors group, the median number of days for nucleic-acid negative conversion was 7.0 days (IQR, 5.0-9.0), which was shorter than the non-RAAS inhibitors group ([median (IQR)] 8.0 (6.0-10.0), P < 0.001, Cohen's d = 0.34), the cumulative negative conversion rates at 3-8 days being all higher than non-RAAS inhibitors groups (P < 0.05). The most significant difference in negative conversion rate between the RAAS inhibitor and non-RAAS inhibitor group was on the 4th day. No significant difference was observed in mean and minimum CT-values from RAAS inhibitor and non-RAAS inhibitor groups (P > 0.05). RAAS inhibitor use in patients with hypertension is associated with nucleic-acid negative conversion duration and negative conversion rate. RAAS inhibitors clearly do not aggravate or prolong COVID-19.
Collapse
Affiliation(s)
- Ru Wen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Medical Imaging, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Jinwen Li
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Fengxi Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian Liu
- Department of Medical Imaging, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Peng Xu
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Mengfei Li
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jingwei Li
- Department of Cardiology, Xinqiao Hospital, Army Military Medical University, Chongqing, China.
- The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia.
| | - Liang Tan
- Department of Critical Care Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
| | - Chen Liu
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
32
|
Greene KS, Choi A, Yang N, Chen M, Li R, Qiu Y, Ezzatpour S, Rojas KS, Shen J, Wilson KF, Katt WP, Aguilar HC, Lukey MJ, Whittaker GR, Cerione RA. Glutamine metabolism is essential for coronavirus replication in host cells and in mice. J Biol Chem 2025; 301:108063. [PMID: 39662828 PMCID: PMC11750454 DOI: 10.1016/j.jbc.2024.108063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/21/2024] [Accepted: 11/30/2024] [Indexed: 12/13/2024] Open
Abstract
Understanding the fundamental biochemical and metabolic requirements for the replication of coronaviruses within infected cells is of notable interest for the development of broad-based therapeutic strategies, given the likelihood of the emergence of new pandemic-potential virus species, as well as future variants of SARS-CoV-2. Here we demonstrate members of the glutaminase family of enzymes (GLS and GLS2), which catalyze the hydrolysis of glutamine to glutamate (i.e., the first step in glutamine metabolism), play key roles in coronavirus replication in host cells. Using a range of human seasonal and zoonotic coronaviruses, we show three examples where GLS expression increases during coronavirus infection of host cells, and another where GLS2 is upregulated. The viruses hijack the metabolic machinery responsible for glutamine metabolism to generate the building blocks for biosynthetic processes and satisfy the bioenergetic requirements demanded by the "glutamine addiction" of virus-infected cells. We demonstrate that genetic silencing of glutaminase enzymes reduces coronavirus infection and that newer members of two classes of allosteric inhibitors targeting these enzymes, designated as SU1, a pan-GLS/GLS2 inhibitor, and UP4, a specific GLS inhibitor, block viral replication in epithelial cells. Moreover, treatment of SARS-CoV-2 infected K18-human ACE2 transgenic mice with SU1 resulted in their complete survival compared to untreated control animals, which succumbed within 10 days post-infection. Overall, these findings highlight the importance of glutamine metabolism for coronavirus replication in human cells and mice and show that glutaminase inhibitors can block coronavirus infection and thereby may represent a novel class of broad-based anti-viral drug candidates.
Collapse
Affiliation(s)
- Kai Su Greene
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Annette Choi
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| | - Nianhui Yang
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Matthew Chen
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Ruizhi Li
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Yijian Qiu
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Shahrzad Ezzatpour
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| | - Katherine S Rojas
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Jonathan Shen
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Kristin F Wilson
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - William P Katt
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Hector C Aguilar
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| | - Michael J Lukey
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Gary R Whittaker
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA; Public & Ecosystem Health, Cornell University, Ithaca, New York, USA
| | - Richard A Cerione
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, USA.
| |
Collapse
|
33
|
Kal M, Brzdęk M, Karska-Basta I, Rzymski P, Pinna A, Mackiewicz J, Odrobina D, Zarębska-Michaluk D, Flisiak R. Difference in Optical Coherence Tomography Angiography Parameters After SARS-CoV-2 Infection During the Alpha and Delta Variant Dominance Periods. Viruses 2024; 17:47. [PMID: 39861835 PMCID: PMC11769401 DOI: 10.3390/v17010047] [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: 12/09/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
The SARS-CoV-2 infection manifests with diverse clinical manifestations, with severity potentially influenced by the viral variant. COVID-19 has also been shown to impact ocular microcirculation in some patients, but whether this effect varies by viral lineage remains unclear. This prospective study compared clinical features and ocular parameters assessed via optical coherence tomography angiography (OCTA) in patients recovering from SARS-CoV-2 infections during the dominance of two distinctive viral lineages, Alpha (B.1.1.7) and Delta (B.1.617.2), and compared them to a control group. The following parameters were measured: vessel density (VD) in the superficial capillary plexus (SCP), deep capillary plexus (DCP), and choriocapillaris (CCP) using OCTA, with a manual assessment of the foveal avascular zones in the SCP (FAZs) and DCP (FAZd). A control group was also included. Among 63 patients in the Alpha group and 41 in the Delta group, no eye-related symptoms were reported during the examination. However, the Delta group showed significantly lower VD in the SCP and DCP across all quadrants (p < 0.001-0.039), while the Alpha group showed reduced VD in the foveal CCP (p = 0.005) and significantly wider FAZs and FAZd (p = 0.002 for both). In conclusion, ocular microcirculatory changes differed between the two variants, with Alpha associated with foveal choroidal VD reduction and larger FAZs and Delta linked to lower SCP and DCP VD across multiple regions. These findings highlight the potential for SARS-CoV-2 variants to differentially impact ocular vasculature, underscoring the need for variant-specific follow-up in COVID-19 patients.
Collapse
Affiliation(s)
- Magdalena Kal
- Collegium Medicum, Jan Kochanowski University, 25-317 Kielce, Poland
- Ophthalmic Clinic, The Voivodeship Hospital, 25-736 Kielce, Poland
| | - Michał Brzdęk
- Collegium Medicum, Jan Kochanowski University, 25-317 Kielce, Poland
- Department of Gastroenterology, Medical University of Lodz, 92-213 Lodz, Poland
| | - Izabella Karska-Basta
- Department of Ophthalmology, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland
- Clinic of Ophthalmology and Ocular Oncology, University Hospital, 31-501 Krakow, Poland
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznan’ University of Medical Sciences, 60-806 Poznan, Poland;
| | - Antonio Pinna
- Department of Medicine, Surgery, and Pharmacy, Ophthalmology Unit, University of Sassari, 07100 Sassari, Italy;
| | - Jerzy Mackiewicz
- Department of Vitreoretinal Surgery, Medical University of Lublin, 20-079 Lublin, Poland
| | - Dominik Odrobina
- Ophthalmic Clinic, The Voivodeship Hospital, 25-736 Kielce, Poland
- Institute of Medical Science, Jan Kochanowski University, 25-317 Kielce, Poland
| | - Dorota Zarębska-Michaluk
- Department of Infectious Diseases and Allergology, Jan Kochanowski University, 25-317 Kielce, Poland
| | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, 15-540 Białystok, Poland;
| |
Collapse
|
34
|
Kudliński B, Zawadzki J, Kulińska W, Kania J, Murkos M, Stolińska M, Zgoła D, Noga A, Nowak P. Tocilizumab in COVID-19: A Double-Edged Sword? Biomedicines 2024; 12:2924. [PMID: 39767830 PMCID: PMC11672960 DOI: 10.3390/biomedicines12122924] [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: 11/05/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: SARS-CoV-2 was responsible for the global pandemic. Approximately 10-15% of patients with COVID-19 developed respiratory failure with adult acute respiratory distress syndrome (ARDS), which required treatment in the Intensive Care Unit (ICU). The cytokine storm observed in severe COVID-19 was frequently handled with steroids. Synergically, tocilizumab, an anti-interleukin-6 receptor monoclonal antibody, gained popularity as a cytokine storm-suppressing agent. However, immunosuppression was proven to increase the predisposition to infections with resistant bacteria. Our study aimed to assess the relationship between positive tests for secondary infections and the survival of patients with severe COVID-19-attributed ARDS treated with immunosuppressive agents. Methods: This study included 342 patients qualified for the ICU and mechanical ventilation (MV). The patients were divided based on the type of immunomodulating therapy and the culture tests results. Results: The results showed the highest survival rate among patients <61 years, favoring the combined treatment (tocilizumab + steroids). Atrial fibrillation (AF) and coronary heart disease (CHD) correlated with a lower survival rate than other comorbidities. Tocilizumab was associated with an increased risk of positive pathogen cultures, which could potentially cause secondary infections; however, the survival rate among these patients was higher. Conclusions: MV and ICU procedures as well as the application of tocilizumab significantly decreased the mortality rate in patients with severe COVID-19-related ARDS. The suppression of cytokine storms played a crucial role in survival. Tocilizumab was found to be both efficient and safe despite the 'side effect' of the increased risk of positive results for secondary infections.
Collapse
Affiliation(s)
- Bartosz Kudliński
- Department of Anesthesia, Critical Care and Rescue Medicine, Collegium Medicum, University in Zielona Góra, 65-046 Zielona Góra, Poland; (J.Z.); (W.K.); (J.K.); (M.M.); (M.S.); (D.Z.); (A.N.); (P.N.)
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gawande V, Kushwaha R, Mandal AA, Banerjee S. Targeting SARS-CoV-2 Proteins: In Silico Investigation with Polypyridyl-Based Zn(II)-Curcumin Complexes. Chembiochem 2024; 25:e202400612. [PMID: 39264259 DOI: 10.1002/cbic.202400612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/13/2024]
Abstract
Herein, we have selected eight Zn(II)-based complexes viz., [Zn(bpy)(acac)Cl] (1), [Zn(phen)(acac)Cl] (2), [Zn(dppz)(acac)Cl] (3), [Zn(dppn)(acac)Cl] (4), [Zn(bpy)(cur)Cl] (5), [Zn(phen)(cur)Cl] (6), [Zn(dppz)(cur)Cl] (7), [Zn(dppn)(cur)Cl] (8), where bpy=2,2'-bipyridine, phen=1,10-phenanthroline, dppz=benzo[i]dipyrido[3,2-a:2',3'-c]phenazine, dppn=naphtho[2,3-i]dipyrido[3,2-a:2',3'-c]phenazine, acac=acetylacetonate, cur=curcumin and performed in silico molecular docking studies with the viral proteins, i. e., spike protein (S), Angiotensin-converting enzyme II Receptor protein (ACE2), nucleocapsid protein (N), main protease protein (Mpro), and RNA-dependent RNA polymerase protein (RdRp) of SARS-CoV-2. The binding energy calculations, visualization of the docking orientation, and analysis of the interactions revealed that these complexes could be potential inhibitors of the viral proteins. Among complexes 1-8, complex 6 showed the strongest binding affinity with S and ACE2 proteins. 4 exerted better binding affinity in the case of the N protein, whereas 8 presented the highest binding affinities with Mpro and RdRp among all the complexes. Overall, the study indicated that Zn(II) complexes have the potential as alternative and viable therapeutic solutions for COVID-19.
Collapse
Affiliation(s)
- Vedant Gawande
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Arif Ali Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| |
Collapse
|
36
|
Oliveira KX, Suzuki YJ. Angiotensin peptides enhance SARS-CoV-2 spike protein binding to its host cell receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628247. [PMID: 39713294 PMCID: PMC11661167 DOI: 10.1101/2024.12.12.628247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the virus that caused the Coronavirus Disease 2019 (COVID-19) pandemic, has a spike glycoprotein that is involved in recognizing and fusing to host cell receptors, such as angiotensin-converting enzyme 2 (ACE2), neuropilin-1 (NRP1), and AXL tyrosine-protein kinase. Since the major spike protein receptor is ACE2, an enzyme that regulates angiotensin II (1-8), this study tested the hypothesis that angiotensin II (1-8) influences the binding of the spike protein to its receptors. While angiotensin II (1-8) did not influence spike-ACE2 binding, we found that it significantly enhances spike-AXL binding. Our experiments showed that longer lengths of angiotensin, such as angiotensin I (1-10), did not significantly affect spike-AXL binding. In contrast, shorter lengths of angiotensin peptides, in particular, angiotensin IV (3-8), strongly increased spike-AXL binding. Angiotensin IV (3-8) also enhanced spike protein binding to ACE2 and NRP1. The discovery of the enhancing effects of angiotensin peptides on spike-host cell receptor binding may suggest that these peptides could be pharmacological targets to treat COVID-19 and post-acute sequelae of SARS-CoV-2 (PASC), which is also known as long COVID.
Collapse
|
37
|
Rong Z, Mai H, Ebert G, Kapoor S, Puelles VG, Czogalla J, Hu S, Su J, Prtvar D, Singh I, Schädler J, Delbridge C, Steinke H, Frenzel H, Schmidt K, Braun C, Bruch G, Ruf V, Ali M, Sühs KW, Nemati M, Hopfner F, Ulukaya S, Jeridi D, Mistretta D, Caliskan ÖS, Wettengel JM, Cherif F, Kolabas ZI, Molbay M, Horvath I, Zhao S, Krahmer N, Yildirim AÖ, Ussar S, Herms J, Huber TB, Tahirovic S, Schwarzmaier SM, Plesnila N, Höglinger G, Ondruschka B, Bechmann I, Protzer U, Elsner M, Bhatia HS, Hellal F, Ertürk A. Persistence of spike protein at the skull-meninges-brain axis may contribute to the neurological sequelae of COVID-19. Cell Host Microbe 2024; 32:2112-2130.e10. [PMID: 39615487 DOI: 10.1016/j.chom.2024.11.007] [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: 08/26/2024] [Revised: 10/10/2024] [Accepted: 11/08/2024] [Indexed: 12/14/2024]
Abstract
SARS-CoV-2 infection is associated with long-lasting neurological symptoms, although the underlying mechanisms remain unclear. Using optical clearing and imaging, we observed the accumulation of SARS-CoV-2 spike protein in the skull-meninges-brain axis of human COVID-19 patients, persisting long after viral clearance. Further, biomarkers of neurodegeneration were elevated in the cerebrospinal fluid from long COVID patients, and proteomic analysis of human skull, meninges, and brain samples revealed dysregulated inflammatory pathways and neurodegeneration-associated changes. Similar distribution patterns of the spike protein were observed in SARS-CoV-2-infected mice. Injection of spike protein alone was sufficient to induce neuroinflammation, proteome changes in the skull-meninges-brain axis, anxiety-like behavior, and exacerbated outcomes in mouse models of stroke and traumatic brain injury. Vaccination reduced but did not eliminate spike protein accumulation after infection in mice. Our findings suggest persistent spike protein at the brain borders may contribute to lasting neurological sequelae of COVID-19.
Collapse
Affiliation(s)
- Zhouyi Rong
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Medical Research School (MMRS), Munich, Germany
| | - Hongcheng Mai
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany; Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Munich Medical Research School (MMRS), Munich, Germany
| | - Gregor Ebert
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Saketh Kapoor
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Jan Czogalla
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Senbin Hu
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jinpeng Su
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Danilo Prtvar
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Inderjeet Singh
- Research Unit Adipocytes & Metabolism (ADM), Helmholtz Diabetes Center, Helmholtz Munich, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Julia Schädler
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claire Delbridge
- Institute of Pathology, Division of Neuropathology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Hanno Steinke
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Hannah Frenzel
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Katja Schmidt
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Christian Braun
- Institute of Legal Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Gina Bruch
- Institute of Legal Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Viktoria Ruf
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mayar Ali
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Graduate School of Neuroscience (GSN), Munich, Germany
| | | | - Mojtaba Nemati
- Department of Neurology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Franziska Hopfner
- Department of Neurology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Selin Ulukaya
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany
| | - Denise Jeridi
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany
| | - Daniele Mistretta
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | | | | | - Fatma Cherif
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Zeynep Ilgin Kolabas
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany; Graduate School of Neuroscience (GSN), Munich, Germany
| | - Müge Molbay
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Medical Research School (MMRS), Munich, Germany
| | - Izabela Horvath
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Center of Doctoral Studies in Informatics and its Applications (CEDOSIA), Technical University of Munich, Munich, Germany
| | - Shan Zhao
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Natalie Krahmer
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Ali Önder Yildirim
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Siegfried Ussar
- Research Unit Adipocytes & Metabolism (ADM), Helmholtz Diabetes Center, Helmholtz Munich, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Susanne M Schwarzmaier
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Benjamin Ondruschka
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Markus Elsner
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany
| | - Harsharan Singh Bhatia
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Farida Hellal
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Koç University, School of Medicine, İstanbul, Turkey.
| |
Collapse
|
38
|
Somova M, Simm S, Ehrhardt J, Schoon J, Burchardt M, Pinto PC. SARS-CoV-2 Spike Protein Amplifies the Immunogenicity of Healthy Renal Epithelium in the Presence of Renal Cell Carcinoma. Cells 2024; 13:2038. [PMID: 39768130 PMCID: PMC11674446 DOI: 10.3390/cells13242038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Renal cell carcinoma (RCC) is the most common form of kidney cancer, known for its immune evasion and resistance to chemotherapy. Evidence indicates that the SARS-CoV-2 virus may worsen outcomes for RCC patients, as well as patients with diminished renal function. Evidence suggests that the SARS-CoV-2 virus may exacerbate outcomes in RCC patients and those with impaired renal function. This study explored the unidirectional effects of RCC cells and the SARS-CoV-2 spike protein (S protein) on human renal proximal tubule epithelial cells (RPTECs) using a microphysiological approach. We co-cultured RCC cells (Caki-1) with RPTEC and exposed them to the SARS-CoV-2 S protein under dynamic 3D conditions. The impact on metabolic activity, gene expression, immune secretions, and S protein internalization was evaluated. The SARS-CoV-2 S protein was internalized by RPTEC but poorly interacted with RCC cells. RPTECs exposed to RCC cells and the S protein exhibited upregulated expression of genes involved in immunogenic pathways, particularly those related to antigen processing and presentation via the major histocompatibility complex I (MHCI). Additionally, increased TNF-α secretion suggested a pro-inflammatory response. Metabolic shifts toward glycolysis were observed in RCC co-culture, while the presence of the S protein led to minor changes. The presence of RCC cells amplified the immune-modulatory effects of the SARS-CoV-2 S protein on the renal epithelium, potentially exacerbating renal inflammation and fostering tumor-supportive conditions. These findings suggest that COVID-19 infections can impact renal function in the presence of kidney cancer.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/virology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
- Kidney Neoplasms/immunology
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Kidney Neoplasms/virology
- SARS-CoV-2/immunology
- COVID-19/immunology
- COVID-19/virology
- COVID-19/metabolism
- Epithelial Cells/metabolism
- Epithelial Cells/virology
- Epithelial Cells/immunology
- Kidney Tubules, Proximal/immunology
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Cell Line, Tumor
- Epithelium/metabolism
- Epithelium/virology
- Epithelium/pathology
- Coculture Techniques
- Kidney/pathology
- Kidney/virology
- Kidney/immunology
- Kidney/metabolism
Collapse
Affiliation(s)
- Maryna Somova
- Department of Urology, University Medicine Greifswald, DZ7 J05.15, Fleischmannstraße 8, 17475 Greifswald, Germany
| | - Stefan Simm
- Institute of Bioinformatics, University Medicine Greifswald, Fleischmannstraße 8, 17475 Greifswald, Germany
- Institute for Bioanalysis, Coburg University of Applied Sciences and Arts, Friedrich-Streib-Str. 2, 96450 Coburg, Germany
| | - Jens Ehrhardt
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Fleischmannstraße 8, 17475 Greifswald, Germany
| | - Janosch Schoon
- Center for Orthopedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, Fleischmannstraße 8, 17475 Greifswald, Germany
| | - Martin Burchardt
- Department of Urology, University Medicine Greifswald, DZ7 J05.15, Fleischmannstraße 8, 17475 Greifswald, Germany
| | - Pedro Caetano Pinto
- Department of Urology, University Medicine Greifswald, DZ7 J05.15, Fleischmannstraße 8, 17475 Greifswald, Germany
| |
Collapse
|
39
|
Minigulov N, Boranbayev K, Bekbossynova A, Gadilgereyeva B, Filchakova O. Structural proteins of human coronaviruses: what makes them different? Front Cell Infect Microbiol 2024; 14:1458383. [PMID: 39711780 PMCID: PMC11659265 DOI: 10.3389/fcimb.2024.1458383] [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: 07/04/2024] [Accepted: 10/17/2024] [Indexed: 12/24/2024] Open
Abstract
Following COVID-19 outbreak with its unprecedented effect on the entire world, the interest to the coronaviruses increased. The causative agent of the COVID-19, severe acute respiratory syndrome coronavirus - 2 (SARS-CoV-2) is one of seven coronaviruses that is pathogenic to humans. Others include SARS-CoV, MERS-CoV, HCoV-HKU1, HCoV-OC43, HCoV-NL63 and HCoV-229E. The viruses differ in their pathogenicity. SARS-CoV, MERS-CoV, and SARS-CoV-2 are capable to spread rapidly and cause epidemic, while HCoV-HKU1, HCoV-OC43, HCoV-NL63 and HCoV-229E cause mild respiratory disease. The difference in the viral behavior is due to structural and functional differences. All seven human coronaviruses possess four structural proteins: spike, envelope, membrane, and nucleocapsid. Spike protein with its receptor binding domain is crucial for the entry to the host cell, where different receptors on the host cell are recruited by different viruses. Envelope protein plays important role in viral assembly, and following cellular entry, contributes to immune response. Membrane protein is an abundant viral protein, contributing to the assembly and pathogenicity of the virus. Nucleocapsid protein encompasses the viral RNA into ribonucleocapsid, playing important role in viral replication. The present review provides detailed summary of structural and functional characteristics of structural proteins from seven human coronaviruses, and could serve as a practical reference when pathogenic human coronaviruses are compared, and novel treatments are proposed.
Collapse
Affiliation(s)
| | | | | | | | - Olena Filchakova
- Biology Department, School of Sciences and Humanities, Nazarbayev
University, Astana, Kazakhstan
| |
Collapse
|
40
|
Ullah MN, Rowan NR, Lane AP. Neuroimmune interactions in the olfactory epithelium: maintaining a sensory organ at an immune barrier interface. Trends Immunol 2024; 45:987-1000. [PMID: 39550314 PMCID: PMC11624989 DOI: 10.1016/j.it.2024.10.005] [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: 09/06/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/18/2024]
Abstract
While primarily a sensory organ, the mammalian olfactory epithelium (OE) also plays a critical role as an immune barrier. Mechanisms governing interactions between the immune system and this specialized chemosensory tissue are gaining interest, in part sparked by the COVID-19 pandemic. Regulated inflammation is intrinsic to normal mucosal healing and homeostasis, but prolonged OE inflammation is associated with persistent loss of smell, belying the intertwining of local mucosal immunology and olfactory function. Evidence supports bidirectional communication between OE cells and the immune system in health and disease. Recent investigations suggest that neuro-immune cross-talk modulates olfactory stem cell behavior and neuronal regeneration dynamics, prioritizing the epithelial-like non-neuronal framework with immune barrier function at the expense of the neurosensory organ in chronic inflammation.
Collapse
Affiliation(s)
- Mohammed N Ullah
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas R Rowan
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew P Lane
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
41
|
Jose J, Ndang K, Chethana MB, Chinmayi CS, Afrana K, Gopan G, Parambi DGT, Munjal K, Chopra H, Dhyani A, Kamal MA. Opportunities and Regulatory Challenges of Functional Foods and
Nutraceuticals During COVID-19 Pandemic. CURRENT NUTRITION & FOOD SCIENCE 2024; 20:1252-1271. [DOI: 10.2174/0115734013276165231129102513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 01/04/2025]
Abstract
:
The novel Coronavirus has brought global mortality, disruption, and a significant loss
of life. A compromised immune system is a known risk factor for all viral influenza infections.
Due to the perceived “immune-boosting” properties of nutraceutical products, sales of dietary supplements have grown globally. In recent years, consumers have increasingly demanded nutraceutical products rather than curative synthetic medicines for preventive therapies for the coronavirus
disease outbreak of 2019 (COVID-19). Healthy foods and nutraceuticals have become daily diet
plans for consumers. Although there has been an increase in demand, there is no such regulation
and harmonized process, which stands as a barrier to the approval of these products. Therefore,
many misbranded and spurious products are entering the market, which may harm consumers.
This article focuses on the role of functional foods and nutraceutical in the management of
COVID-19 also focuses on the different nutraceutical regulations in each country and compare the
similarities and differences of the following countries: India, the USA (United States of America),
the EU (European Union), and China. The comparative study of nutraceutical regulations in India,
the USA, Europe, and China shows that there is a difference regarding the nutraceutical regulations; however, despite the differences, it is observed that it has the same underlying objective,
i.e., ensuring the safety of the consumers by maintaining the product quality.
Collapse
Affiliation(s)
- Jobin Jose
- Department of Pharmaceutics and Pharmaceutical Regulatory Affairs, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore 575018, India
| | - Keyidaule Ndang
- Department of Pharmaceutics and Pharmaceutical Regulatory Affairs, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore 575018, India
| | - Madhusoodhana Ballakkuraya Chethana
- Department of Pharmaceutics and Pharmaceutical Regulatory Affairs, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore 575018, India
| | - Chikmagalur Srinath Chinmayi
- Department of Pharmaceutics and Pharmaceutical Regulatory Affairs, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore 575018, India
| | - Khatheeja Afrana
- Department of Pharmaceutics and Pharmaceutical Regulatory Affairs, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore 575018, India
| | - Gopika Gopan
- Department of Pharmaceutics and Pharmaceutical Regulatory Affairs, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore 575018, India
| | - Della Grace Thomas Parambi
- Department of Pharmaceutical Chemistry, College of
Pharmacy, Jouf University, Sakaka, Al Jouf 72341, Saudi Arabia
| | - Kavita Munjal
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering,
Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, Tamil Nadu, India
| | - Archana Dhyani
- School of Pharmacy,
Graphic Era Hill University, Dehradun, India
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-
related Molecular Network, West China Hospital, Sichuan University, Sichuan, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health
Sciences, Daffodil International University, Dhaka, Bangladesh
- Enzymoics, 7 Peterlee place, Hebersham, NSW
2770; Novel Global Community Educational Foundation, Australia
| |
Collapse
|
42
|
Fajar JK, Tamara F, Putranto W, Prabowo NA, Harapan H. Insertion/deletion (I/D) polymorphisms of angiotensin-converting enzyme gene and their implications for susceptibility and severity of COVID-19: A systematic review and meta-analysis. NARRA J 2024; 4:e727. [PMID: 39816082 PMCID: PMC11731805 DOI: 10.52225/narra.v4i3.727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 12/12/2024] [Indexed: 01/18/2025]
Abstract
The insertion or deletion polymorphisms of the angiotensin-converting enzyme gene (ACE I/D) have been the subject of significant research related to coronavirus disease 2019 (COVID-19). Despite this, the findings have remained uncertain and debatable. The aim of this study was to determine the associations between the ACE I/D polymorphisms and the susceptibility as well as the severity of COVID-19. A meta-analysis study (PROSPERO: CRD42022384562) was conducted by searching the articles published on PubMed, Scopus, and Embase as of May 15, 2023. Information regarding the impact of ACE I/D variant on the susceptibility to COVID-19 and its severity was collected and analyzed utilizing the Mantel-Haenszel method with a random effects model or fixed effects model, depending on the presence or absence of heterogeneity. Out of 3,335 articles, 21 articles were included, of which 13 investigated the association between ACE I/D and the risk of COVID-19 infection and 18 of them examined its influence on disease severity. The D allele of ACE increased risk of COVID-19 infection (OR: 1.41; 95%CI: 1.08-1.85; p-Egger: 0.0676; p-Heterogeneity: <0.001; p=0.0120), while ACE I allele (OR: 0.71; 95%CI: 0.54-0.93; p-Egger: 0.0676; p-Heterogeneity: <0.001; p=0.012) and II genotype (OR: 0.55; 95%CI: 0.34-0.87; p-Egger: 0.200; p-Heterogeneity: <0.001; p=0.011) decreased the risk of infection. Additionally, there was a notable association between the ACE ID genotype and an elevated likelihood of experiencing severe COVID-19 within the Asian population (OR: 1.46; 95%CI: 1.15-1.84; p-Egger: 0.092; p-Heterogeneity: 0.116; p=0.002). The presence of ACE I/D polymorphisms significantly influences the likelihood of being susceptible to and experiencing the severity of COVID-19.
Collapse
Affiliation(s)
- Jonny K. Fajar
- Department of Internal Medicine, Rumah Sakit Universitas Brawijaya, Malang, Indonesia
| | - Fredo Tamara
- Division of Nephrology and Hypertension, Department of Internal Medicine, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Wachid Putranto
- Division of Nephrology and Hypertension, Department of Internal Medicine, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Nurhasan A. Prabowo
- Department of Internal Medicine, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Harapan Harapan
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| |
Collapse
|
43
|
Pavuluri P, Menon MG, Tummalacharla SC, Sameer Raheem S, Karpay S, Chepuri P. Liver Enzymes and Inflammatory Markers Among Severely Ill COVID-19 Patients: A Retrospective Case-Control Study in Telangana. Cureus 2024; 16:e75120. [PMID: 39759701 PMCID: PMC11698693 DOI: 10.7759/cureus.75120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Introduction The COVID-19 pandemic originated in Wuhan, China, and swiftly spread across all continents. The respiratory system is the most affected in people who acquire sickness as a result of SARS-CoV-2. However, the virus can also affect other systems. The COVID-19 pandemic has become one of the most fatal infectious diseases in the recent past. Patients present with symptoms of fever, cough, tiredness, loss of taste or smell, sore throat, headache, and diarrhea. Objective This study intends to evaluate how COVID-19 has shown its effects on the well-being of the liver by collecting and correlating the data of the liver enzymes and inflammatory markers among hospitalized COVID-19 patients with age- and sex-matched healthy controls. Materials and methods A retrospective case-control study that included 200 patients diagnosed and hospitalized with COVID-19 was compared with an equal number of age- and sex-matched healthy control groups without COVID-19 at RVM Institute of Medical Sciences and Research Centre (RVMIMS & RC), a tertiary care teaching hospital in Siddipet, Telangana, India. Liver function tests (LFTs) and inflammatory markers were evaluated in both groups. Results Out of 200 patients, 179 (89.5%) had elevated alanine transaminase (ALT), 191 (95.5%) had elevated aspartate aminotransferase (AST), 33 (16.5%) had elevated alkaline phosphatase (ALP), and 183 (91.5%) showed elevated D-dimer levels. All the patients had elevated interleukin-6 (IL-6) and C-reactive protein (CRP) levels. Conclusion COVID-19 patients have exhibited elevations in liver enzyme panels and inflammatory markers. Further research and follow-up studies may aid in understanding the role of the well-being of the liver in patients affected by COVID-19. Considering the emergence of newer COVID-19 strains, we recommend LFT to patients who test positive for the virus to monitor prognosis and guide treatment protocols through this study.
Collapse
Affiliation(s)
- Pratyusha Pavuluri
- Biochemistry, RVM Institute of Medical Sciences and Research Center, Hyderabad, IND
| | - M Girija Menon
- Biochemistry, RVM Institute of Medical Sciences and Research Center, Hyderabad, IND
| | | | - Shaik Sameer Raheem
- Biochemistry, RVM Institute of Medical Sciences and Research Center, Hyderabad, IND
| | - Soujanya Karpay
- Biochemistry, RVM Institute of Medical Sciences and Research Center, Hyderabad, IND
| | - Phanindra Chepuri
- Biochemistry, RVM Institute of Medical Sciences and Research Center, Hyderabad, IND
| |
Collapse
|
44
|
Ghanem L, Essayli D, Kotaich J, Zein MA, Sahebkar A, Eid AH. Phenotypic switch of vascular smooth muscle cells in COVID-19: Role of cholesterol, calcium, and phosphate. J Cell Physiol 2024; 239:e31424. [PMID: 39188012 PMCID: PMC11649971 DOI: 10.1002/jcp.31424] [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: 03/06/2024] [Revised: 07/11/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Although the novel coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), primarily manifests as severe respiratory distress, its impact on the cardiovascular system is also notable. Studies reveal that COVID-19 patients often suffer from certain vascular diseases, partly attributed to increased proliferation or altered phenotype of vascular smooth muscle cells (VSMCs). Although the association between COVID-19 and VSMCs is recognized, the precise mechanism underlying SARS-CoV-2's influence on VSMC phenotype remains largely under-reviewed. In this context, while there is a consistent body of literature dissecting the effect of COVID-19 on the cardiovascular system, few reports delve into the potential role of VSMC switching in the pathophysiology associated with COVID-19 and the molecular mechanisms involved therein. This review dissects and critiques the link between COVID-19 and VSMCs, with particular attention to pathways involving cholesterol, calcium, and phosphate. These pathways underpin the interaction between the virus and VSMCs. Such interaction promotes VSMC proliferation, and eventually potentiates vascular calcification as well as worsens prognosis in patients with COVID-19.
Collapse
MESH Headings
- Animals
- Humans
- Calcium/metabolism
- Cell Proliferation
- Cholesterol/metabolism
- COVID-19/metabolism
- COVID-19/pathology
- COVID-19/virology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/virology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/virology
- Phenotype
- Phosphates/metabolism
- SARS-CoV-2/pathogenicity
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Vascular Calcification/virology
Collapse
Affiliation(s)
- Laura Ghanem
- Faculty of Medical SciencesLebanese UniversityHadathLebanon
| | - Dina Essayli
- Faculty of Medical SciencesLebanese UniversityHadathLebanon
| | - Jana Kotaich
- Faculty of Medical SciencesLebanese UniversityHadathLebanon
- MEDICA Research InvestigationBeirutLebanon
| | | | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Biotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU HealthQatar UniversityDohaQatar
| |
Collapse
|
45
|
Raza ML, Imam MH, Zehra W, Jamil S. Neuro-inflammatory pathways in COVID-19-induced central nervous system injury: Implications for prevention and treatment strategies. Exp Neurol 2024; 382:114984. [PMID: 39368535 DOI: 10.1016/j.expneurol.2024.114984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/15/2024] [Accepted: 09/29/2024] [Indexed: 10/07/2024]
Abstract
This review explores the neuroinflammatory pathways underlying COVID-19-induced central nervous system (CNS) injury, with a focus on mechanisms of brain damage and strategies for prevention. A comprehensive literature review was conducted to summarize current knowledge on the pathways by which SARS-CoV-2 reaches the brain, the neuroinflammatory responses triggered by viral infection, neurological symptoms and long COVID. Results: We discuss the mechanisms of neuroinflammation in COVID-19, including blood-brain barrier disruption, cytokine storm, microglial activation, and peripheral immune cell infiltration. Additionally, we highlight potential strategies for preventing CNS injury, including pharmacological interventions, immunomodulatory therapies, and lifestyle modifications. Conclusively, Understanding the neuroinflammatory pathways in COVID-19-induced CNS injury is crucial for developing effective prevention and treatment strategies to protect brain health during and after viral infection.
Collapse
Affiliation(s)
- Muhammad Liaquat Raza
- Department of Infection Prevention & Control, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| | | | | | - Subia Jamil
- Faculty of Pharmacy, Jinnah University for Women, University, Karachi, Pakistan
| |
Collapse
|
46
|
Fallah A, Sedighian H, Kachuei R, Fooladi AAI. Human microbiome in post-acute COVID-19 syndrome (PACS). CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 8:100324. [PMID: 39717208 PMCID: PMC11665312 DOI: 10.1016/j.crmicr.2024.100324] [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] [Indexed: 12/25/2024] Open
Abstract
The global COVID-19 pandemic, which began in 2019, is still ongoing. SARS-CoV-2, also known as the severe acute respiratory syndrome coronavirus 2, is the causative agent. Diarrhea, nausea, and vomiting are common GI symptoms observed in a significant number of COVID-19 patients. Additionally, the respiratory and GI tracts express high level of transmembrane protease serine 2 (TMPRSS2) and angiotensin-converting enzyme-2 (ACE2), making them primary sites for human microbiota and targets for SARS-CoV-2 infection. A growing body of research indicates that individuals with COVID-19 and post-acute COVID-19 syndrome (PACS) exhibit considerable alterations in their microbiome. In various human disorders, including diabetes, obesity, cancer, ulcerative colitis, Crohn's disease, and several viral infections, the microbiota play a significant immunomodulatory role. In this review, we investigate the potential therapeutic implications of the interactions between host microbiota and COVID-19. Microbiota-derived metabolites and components serve as primary mediators of microbiota-host interactions, influencing host immunity. We discuss the various mechanisms through which these metabolites or components produced by the microbiota impact the host's immune response to SARS-CoV-2 infection. Additionally, we address confounding factors in microbiome studies. Finally, we examine and discuss about a range of potential microbiota-based prophylactic measures and treatments for COVID-19 and PACS, as well as their effects on clinical outcomes and disease severity.
Collapse
Affiliation(s)
- Arezoo Fallah
- Department of Bacteriology and Virology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
47
|
Bukowska B. Current and Potential Use of Biologically Active Compounds Derived from Cannabis sativa L. in the Treatment of Selected Diseases. Int J Mol Sci 2024; 25:12738. [PMID: 39684447 DOI: 10.3390/ijms252312738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Cannabis sativa L. contains numerous compounds with antioxidant and anti-inflammatory properties, including the flavonoids and the cannabinoids, particularly Δ-9-tetrahydrocannabinol (THC) and cannabidiol (CBD). Cannabinoids have an effect on the endocannabinoid system (ECS), a cellular communication network, and are, hence, widely studied for medical applications. Epidiolex®, a 99% pure oral CBD extract, has been approved by the FDA for the treatment of epilepsy. Nabiximols (Sativex) is an oromucosal spray containing equal volume of THC and CBD, and it is commonly used as an add-on treatment for unresponsive spasticity in multiple sclerosis (MS) patients. Several in vitro and in vivo studies have also shown that cannabinoids can be used to treat various types of cancer, such as melanoma and brain glioblastoma; the first positive clinical trials on the anticancer effect of a THC:CBD blend with temozolomide (TMZ) in the treatment of highly invasive brain cancer are very promising. The cannabinoids exert their anticancer properties in in vitro investigations by the induction of cell death, mainly by apoptosis and cytotoxic autophagy, and the inhibition of cell proliferation. In several studies, cannabinoids have been found to induce tumor regression and inhibit angiogenic mechanisms in vitro and in vivo, as well as in two low-numbered epidemiological studies. They also exhibit antiviral effects by inhibiting ACE2 transcription, blocking viral replication and fusion, and acting as anti-inflammatory agents; indeed, prior CBD consumption (a study of 93,565 persons in Chicago) has also been associated with a much lower incidence of SARS-CoV-2 infections. It is postulated that cannabis extracts can be used in the treatment of many other diseases such as systemic lupus erythematosus, type 1 diabetes, or various types of neurological disorders, e.g., Alzheimer's disease. The aim of this review is to outline the current state of knowledge regarding currently used medicinal preparations derived from C. sativa L. in the treatment of selected cancer and viral diseases, and to present the latest research on the potential applications of its secondary metabolites.
Collapse
Affiliation(s)
- Bożena Bukowska
- Department of Biophysics of Environmental Pollution, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska Street141/143, 90-236 Lodz, Poland
| |
Collapse
|
48
|
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
|
49
|
Sosnowska M, Wierzbicki M, Nasiłowska B, Bakalova TN, Piotrowska K, Strojny-Cieślak B, Sawosz E, Kutwin M. Fullerenol C 60(OH) 40 Nanoparticles and Ectoine Protect Human Nasal Epithelial Cells Against the Cytokine Storm After Addition of the Full-Length Spike Protein from SARS-CoV-2. Int J Nanomedicine 2024; 19:12221-12255. [PMID: 39600409 PMCID: PMC11588572 DOI: 10.2147/ijn.s482652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/10/2024] [Indexed: 11/29/2024] Open
Abstract
INTRODUCTION AND OBJECTIVE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) enters the nasal cavity, penetrates the nasal epithelial cells through the interaction of its spike protein with the host cell receptor angiotensin-converting enzyme 2 (ACE2) and then triggers a cytokine storm. We aimed to assess the biocompatibility of fullerenol nanoparticles C60(OH)40 and ectoine, and to document their effect on the protection of primary human nasal epithelial cells (HNEpCs) against the effects of interaction with the fragment of virus - spike protein. This preliminary research is the first step towards the construction of a intranasal medical device with a protective, mechanical function against SARS-CoV-2 similar to that of personal protective equipment (eg masks). METHODS We used HNEpCs and the full-length spike protein from SARS-CoV-2 to mimic the first stage of virus infection. We assessed cell viability with the XTT assay and a spectrophotometer. May-Grünwald Giemsa and periodic acid-Schiff staining served to evaluate HNEpC morphology. We assessed reactive oxygen species (ROS) production by using 2',7'-dichlorofluorescin diacetate and commercial kit. Finally, we employed reverse transcription polymerase chain reaction, Western blotting and confocal microscopy to determine the expression of angiotensin-converting enzyme 2 (ACE2) and inflammatory cytokines. RESULTS There was normal morphology and unchanged viability of HNEpCs after incubation with 10 mg/L C60(OH)40, 0.2% ectoine or their composite for 24 h. The spike protein exerted cytotoxicity via ROS production. Preincubation with the composite protected HNEpCs against the interaction between the spike protein and the host membrane and prevented the production of key cytokines characteristic of severe coronavirus disease 2019, including interleukin 6 and 8, monocyte chemotactic protein 1 and 2, tissue inhibitor of metalloproteinases 2 and macrophage colony-stimulating factor. CONCLUSION In the future, the combination of fullerenol and ectoine may be used to prevent viral infections as an intranasal medical device for people with reduced immunity and damaged mucous membrane.
Collapse
Affiliation(s)
- Malwina Sosnowska
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Mateusz Wierzbicki
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Barbara Nasiłowska
- Biomedical Engineering Center, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Totka Nikolaeva Bakalova
- Department of Material Science, Faculty of Mechanical Engineering, Technical University of Liberec, Liberec, Czech Republic
| | - Klara Piotrowska
- Department of Animal Nutrition, Institute of Animal Sciences, Warsaw University of Life Sciences, Warsaw, Poland
| | - Barbara Strojny-Cieślak
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Ewa Sawosz
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Marta Kutwin
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| |
Collapse
|
50
|
Scalzo PL, Marshall A, Soriano S, Curry K, Dulay M, Hodics T, Quigley EMM, Treangen TJ, Piskorz MM, Villapol S. Gut microbiome dysbiosis and immune activation correlate with somatic and neuropsychiatric symptoms in COVID-19 patients: Microbiome dysbiosis linked to COVID-19 symptoms. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.18.24317428. [PMID: 39606341 PMCID: PMC11601728 DOI: 10.1101/2024.11.18.24317428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
COVID-19 patients often exhibit altered immune responses and neuropsychiatric symptoms during hospitalization. However, the potential interactions with gut microbiome profiles have not been fully characterized. Here, COVID-19 disease severity was classified as low (27.4%), moderate (29.8%), and critical (42.8%). Fever (66.1%) and cough (55.6%) were common symptoms. Additionally, 27.3% reported somatic symptoms, 27.3% experienced anxiety, 39% had depressive symptoms, and 80.5% reported stress. Gut microbiome profiling was performed using full-length 16S rRNA gene sequencing. Elevated interleukin-6 levels were observed in the most severe cases, indicating systemic inflammation. Reduced gut bacterial diversity was more pronounced in women and obese patients and correlated with higher disease severity. The presence of the genus Mitsuokella was significantly associated with increased physical, stress, anxiety, and depressive symptoms, and Granulicatella with critically ill patients. These findings suggest a link between mental health status, systemic inflammation, and gut dysbiosis in COVID-19 patients, emphasizing the potential of microbiome-targeted therapies to improve recovery and reduce severe complications.
Collapse
Affiliation(s)
- Paula L. Scalzo
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, MG, Brazil
| | - Austin Marshall
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Computer Science, Rice University, Houston, TX, USA
| | - Sirena Soriano
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Kristen Curry
- Department of Computer Science, Rice University, Houston, TX, USA
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris 75015, France
| | - Mario Dulay
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Timea Hodics
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Eamonn MM Quigley
- Lynda K. and David M. Underwood Center for Digestive Health, Houston Methodist Hospital, Houston, TX, USA
| | - Todd J. Treangen
- Department of Computer Science, Rice University, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - María M. Piskorz
- Department of Neurogastroenterology, Hospital de Clinicas José de San Martin, Universidad de Buenos Aires, Argentina
| | - Sonia Villapol
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Neuroscience in Neurological Surgery, Weill Cornell Medical College, NY, USA
| |
Collapse
|