1
|
Ito T, Suzuki T, Sakai Y, Nishioka K, Itoh Y, Sakamoto K, Ikemura N, Matoba S, Kanda Y, Takagi J, Okamoto T, Tahara K, Hoshino A. Engineered ACE2 decoy in dry powder form for inhalation: A novel therapy for SARS-CoV-2 variants. Mol Ther Methods Clin Dev 2025; 33:101459. [PMID: 40276779 PMCID: PMC12019485 DOI: 10.1016/j.omtm.2025.101459] [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: 11/12/2024] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
The persistent threat of SARS-CoV-2 and the emergence of new variants has prompted the development of a novel, easily administered modality that can overcome viral mutations. The engineered ACE2 decoy shows neutralizing activity comparable to monoclonal antibodies and is broadly effective against SARS-CoV-2 variants and ACE2-utilizing sarbecoviruses. In addition to intravenous administration, this decoy has shown antiviral efficacy through nebulized aerosol inhalation in murine and primate models, offering a dose-sparing advantage. Clinically, dry powder formulation is ideal for convenience and storage but poses challenges for protein biologics. This study developed a freeze-dried spray formulation of the ACE2 decoy for inhalation. The trehalose and leucine-based excipient maintained neutralizing activity and prevented aggregate formation. The dry powder showed aerodynamic distribution from bronchi to alveoli, aiding protection against SARS-CoV-2 infections. Neutralizing activity, structural stability, and powder dispersibility were preserved after 6 months of storage. In a mouse model of SARS-CoV-2 infection, significant reductions in viral replication and lung pathology were observed with intratracheal administration 24 h post-infection. The ACE2 decoy retained activity against recent JN.1 and current KP.3 strains, confirming its robust efficacy against viral mutations. This ACE2 decoy powder inhalant is a self-administered, next-generation treatment addressing the ongoing immune-evading evolution of SARS-CoV-2.
Collapse
Affiliation(s)
- Takaaki Ito
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tatsuya Suzuki
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yusuke Sakai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Keisuke Nishioka
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yumi Itoh
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kentarou Sakamoto
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Nariko Ikemura
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | - Junichi Takagi
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| | - Toru Okamoto
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kohei Tahara
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu 501-1196, Japan
- Laboratory of Nanofiber Technology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Atsushi Hoshino
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
2
|
Bi Z, Ren W, Zeng H, Zhou Y, Liu J, Chen Z, Zhang X, He X, Lu G, Wei Y, Wei X. LL-37 Inhibits TMPRSS2-Mediated S2' Site Cleavage and SARS-CoV-2 Infection but Not Omicron Variants. Cell Prolif 2025:e70060. [PMID: 40375579 DOI: 10.1111/cpr.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/10/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025] Open
Abstract
Continual evolution of SARS-CoV-2 spike drives the emergence of Omicron variants that show increased spreading and immune evasion. Understanding how the variants orientate themselves towards host immune defence is crucial for controlling future pandemics. Herein, we demonstrate that human cathelicidin LL-37, a crucial component of innate immunity, predominantly binds to the S2 subunit of SARS-CoV-2 spike protein, occupying sites where TMPRSS2 typically binds. This binding impedes TMPRSS2-mediated priming at site S2' and subsequent membrane fusion processes. The mutation N764K within S2 subunit of Omicron variants reduces affinity for LL-37 significantly, thereby diminishing binding capacity and inhibitory effects on membrane fusion. Moreover, the early humoral immune response enhanced by LL-37 is observed in mice against SARS-CoV-2 spike but not Omicron BA.4/5 spike. These findings reveal the mechanism underlying interactions amongst LL-37, TMPRSS2 and SARS-CoV-2 and VOCs, and highlight the distinct mutation for Omicron variants to evade the fusion activity inhibition by host innate immunity.
Collapse
Affiliation(s)
- Zhenfei Bi
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wenyan Ren
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanyuan Zhou
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, China
| | - Jian Liu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zimin Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xindan Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Guangwen Lu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Guthrie C, Meeker AC, Self AE, Ramos-Leyva A, Clark OL, Kotey SK, Hartson SD, Liang Y, Liu L, Tan X, Cheng Y. Microvesicles Derived from Human Bronchial Epithelial Cells Regulate Macrophage Activation During Mycobacterium abscessus Infection. J Proteome Res 2025; 24:2291-2301. [PMID: 40153482 PMCID: PMC12053935 DOI: 10.1021/acs.jproteome.4c00827] [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: 09/21/2024] [Revised: 01/26/2025] [Accepted: 03/24/2025] [Indexed: 03/30/2025]
Abstract
Intercellular communication is important for host immunity in response to bacterial infections. Nontuberculous mycobacterium (NTM), such as Mycobacterium abscessus (M. ab), is a group of environmental bacteria that can cause severe lung infections in individuals with pre-existing lung conditions, including cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD). There is limited knowledge understanding the interaction between airway epithelial cells and immune cells during NTM infections. In this study, we characterized microvesicles (MVs) released from uninfected and M. ab-infected human bronchial epithelial cells and investigated the effect of these MVs on the activation and polarization of THP-1-derived macrophages in cell culture. Our results indicate that MVs released by M. ab-infected human bronchial epithelial cells stimulated the activation of M2-polarized macrophages in cell culture when compared to MVs released by uninfected cells. Additionally, the proteomic analysis for isolated MVs showed that the proteins involved in the cell adhesion pathway were enriched in MVs from M. ab-infected human bronchial epithelial cells compared to MVs from uninfected cells. Among those, the cell surface protein, intercellular adhesion molecule 1 (ICAM-1), regulated the uptake of MVs released by M. ab-infected human bronchial epithelial cells by recipient macrophages in cell culture. In conclusion, our data suggest that in response to M. ab infection, human airway epithelial cells release MVs to modulate the activation of macrophages, which are key cells for mycobacterial intracellular survival in the host.
Collapse
Affiliation(s)
- Carlyn
M. Guthrie
- Department
of Biochemistry and Molecular Biology, Oklahoma
State University, Stillwater, Oklahoma 74078, United States
- Oklahoma
Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Amber C. Meeker
- Department
of Biochemistry and Molecular Biology, Oklahoma
State University, Stillwater, Oklahoma 74078, United States
- Oklahoma
Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Ashton E. Self
- Department
of Biochemistry and Molecular Biology, Oklahoma
State University, Stillwater, Oklahoma 74078, United States
- Oklahoma
Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Aidaly Ramos-Leyva
- Department
of Biochemistry and Molecular Biology, Oklahoma
State University, Stillwater, Oklahoma 74078, United States
- Oklahoma
Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Olivia L. Clark
- Department
of Biochemistry and Molecular Biology, Oklahoma
State University, Stillwater, Oklahoma 74078, United States
- Oklahoma
Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Stephen K. Kotey
- Department
of Biochemistry and Molecular Biology, Oklahoma
State University, Stillwater, Oklahoma 74078, United States
- Oklahoma
Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Steven D. Hartson
- Department
of Biochemistry and Molecular Biology, Oklahoma
State University, Stillwater, Oklahoma 74078, United States
- Center
for Genomics and Proteomics, Oklahoma State
University, Stillwater, Oklahoma 74078, United States
| | - Yurong Liang
- Oklahoma
Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma 74078, United States
- Department
of Physiological Sciences, Oklahoma State
University, Stillwater, Oklahoma 74078, United States
| | - Lin Liu
- Oklahoma
Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma 74078, United States
- Department
of Physiological Sciences, Oklahoma State
University, Stillwater, Oklahoma 74078, United States
| | - Xuejuan Tan
- Department
of Biochemistry and Molecular Biology, Oklahoma
State University, Stillwater, Oklahoma 74078, United States
- Oklahoma
Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Yong Cheng
- Department
of Biochemistry and Molecular Biology, Oklahoma
State University, Stillwater, Oklahoma 74078, United States
- Oklahoma
Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| |
Collapse
|
4
|
Park DE, Aziz M, Salazar JE, Pham T, Nelson SG, Villani J, Weber NO, Price LB, Hungate BA, Liu CM. The nasal microbiome modulates risk for SARS-CoV-2 infection. EBioMedicine 2025; 115:105660. [PMID: 40210576 DOI: 10.1016/j.ebiom.2025.105660] [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: 10/22/2024] [Revised: 02/20/2025] [Accepted: 03/06/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND The nasal microbiome may influence host risk for COVID-19 by modulating the expression of key proteins that facilitate SARS-CoV-2 entry, including angiotensin-converting enzyme 2 (ACE2), which binds the virus, and transmembrane serine protease 2 (TMPRSS2), which activates viral entry into nasal epithelial cells. This study examined whether the expression levels of ACE2 and TMPRSS2 in the nasal cavity predict the risk of SARS-CoV-2 infection and whether the host nasal microbiome modulates their expression. METHODS Using 1548 self-collected nasal swabs from a population-based surveillance testing of community-dwelling adults in Washington D.C., we conducted two retrospective case-control studies (cross-sectional: n = 111 cases and 343 controls; longitudinal: n = 97 cases, 286 controls) and a nasal microbiome study (n = 428). Cases, defined as individuals with a positive SARS-CoV-2 test, were matched with controls based on age and test date. Pre-infection samples were analysed. We measured nasal ACE2/TMPRSS2 expression using RT-qPCR and characterized the nasal microbiome using 16S rRNA gene-based qPCR and sequencing. We used machine learning and regression analysis to determine if nasal ACE2/TMPRSS2 expression predicts SARS-CoV-2 infection and whether the nasal microbiome influences their expression. FINDINGS Elevated nasal ACE2/TMPRSS2 expression was associated with 3.6-fold increased risk of contracting COVID-19 (95% CI = 1.71-7.47) compared to those with no detectable levels of ACE2 or TMPRSS2. Before testing positive for SARS-CoV-2, cases also had significantly higher and more unstable ACE2/TMPRSS2 expression in their nasal cavity than controls. Having high densities of Staphylococcus aureus, Haemophilus influenzae, or Moraxella catarrhalis/nonliquefaciens was linked to increased nasal ACE2/TMPRSS2 expression. In contrast, having high densities of Dolosigranulum pigrum was associated with decreased nasal ACE2/TMPRSS2 expression. INTERPRETATION These results suggest that natural variation in the nasal microbiome significantly impacts ACE2/TMPRSS2 expression in the nasal cavity and the near-term risk of SARS-CoV-2 infection in adults. Modifying the nasal microbiome could potentially reduce COVID-19 risk. FUNDING Research reported in this article was supported by the Milken Institute School of Public Health, the George Washington University and the National Institute of Allergy and Infectious Diseases, National Institutes of Health under award number R01AI168182. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Collapse
Affiliation(s)
- Daniel E Park
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Maliha Aziz
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Juan E Salazar
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Tony Pham
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Sydney G Nelson
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Jack Villani
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Nathan O Weber
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Lance B Price
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Bruce A Hungate
- Center for Ecosystem Science and Society, Northern Arizona University, Flagstaff, AZ, 86011, USA; Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, 86011, USA
| | - Cindy M Liu
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
5
|
Felix FA, Filiú FMV, da Silva TA, Calderaro DC, Tanure LA, de Castro MAA, Gomez RS, Diniz MG, de Sousa SF. SARS-CoV-2 Entry Factors ACE2 , TMPRSS2 and FURIN in Salivary Glands From Primary Sjögren's Disease. J Oral Pathol Med 2025; 54:290-297. [PMID: 40091593 DOI: 10.1111/jop.13621] [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: 10/29/2024] [Accepted: 01/30/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Salivary glands are the reservoir for severe acute respiratory syndrome Coronavirus 2 virus (SARS-CoV-2). It is known that SARS-CoV-2 uses angiotensin-converting enzyme 2 (ACE2), transmembrane serine protease 2 (TMPRSS2) and FURIN to infect cells. Salivary glands express these entry factors, and it is unclear if these molecules are expressed in salivary glands from primary Sjögren's disease (pSjD). This study assessed ACE2, TMPRSS2, and FURIN gene expression by RT-qPCR in pSjD compared to normal salivary gland and pleomorphic adenoma. METHODS An integrated study of ACE2, TMPRSS2, and FURIN gene expression across pSjD, pleomorphic adenoma, and normal salivary gland by RT-pPCR was performed. The association between gene expression and clinicopathological data was also examined. RESULTS pSjD showed ACE2 upregulation compared with normal salivary gland and pleomorphic adenoma (p < 0.05). TMPRSS2 and FURIN mRNA levels are also higher in pSjD compared to pleomorphic adenoma (p < 0.05). ACE2 expression levels were associated with anti-SSA/SSB positivity (p < 0.05). A strong correlation was observed between TMPRSS2 and FURIN in pSjD (r = 0.768; p = 0.001). CONCLUSION Our analysis revealed a high expression of ACE2 in pSjD. ACE2, TMPRSS2, and FURIN mRNA levels are increased in pSjD in comparison to pleomorphic adenomas. Neither ACE2, TMPRSS2, nor furin was consistently associated with SARS-CoV-2 positivity in pSjD. These findings may guide future investigations on the effects of SARS-CoV-2 in a subset of salivary gland diseases.
Collapse
Affiliation(s)
- Fernanda Aragão Felix
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Flávia Martins Vasconcelos Filiú
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Tarcília Aparecida da Silva
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Leandro Augusto Tanure
- Hospital das Clínicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maurício Augusto Aquino de Castro
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo Santiago Gomez
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marina Gonçalves Diniz
- Department of Pathology, Biological Science Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sílvia Ferreira de Sousa
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
6
|
de Melo BP, da Silva JAM, Rodrigues MA, Palmeira JDF, Amato AA, Argañaraz GA, Argañaraz ER. SARS-CoV-2 Spike Protein and Long COVID-Part 2: Understanding the Impact of Spike Protein and Cellular Receptor Interactions on the Pathophysiology of Long COVID Syndrome. Viruses 2025; 17:619. [PMID: 40431631 DOI: 10.3390/v17050619] [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: 02/28/2025] [Revised: 04/06/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
SARS-CoV-2 infection has had a significant impact on global health through both acute illness, referred to as coronavirus disease 2019 (COVID-19), and chronic conditions (long COVID or post-acute sequelae of COVID-19, PASC). Despite substantial advancements in preventing severe COVID-19 cases through vaccination, the rise in the prevalence of long COVID syndrome and a notable degree of genomic mutation, primarily in the S protein, underscores the necessity for a deeper understanding of the underlying pathophysiological mechanisms related to the S protein of SARS-CoV-2. In this review, the latest part of this series, we investigate the potential pathophysiological molecular mechanisms triggered by the interaction between the spike protein and cellular receptors. Therefore, this review aims to provide a differential and focused view on the mechanisms potentially activated by the binding of the spike protein to canonical and non-canonical receptors for SARS-CoV-2, together with their possible interactions and effects on the pathogenesis of long COVID.
Collapse
Affiliation(s)
- Bruno Pereira de Melo
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Jhéssica Adriane Mello da Silva
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Mariana Alves Rodrigues
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Julys da Fonseca Palmeira
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Angélica Amorim Amato
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Gustavo Adolfo Argañaraz
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Enrique Roberto Argañaraz
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| |
Collapse
|
7
|
AbdelGhany Morsy SA, Abd El Mottelib LMMA, Assem S, Abd El Aziz MM, Elgeziry AH. Pioglitazone mitigates acetic acid-induced colitis in rats via epigenetic-modulation and antioxidant mechanisms. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04109-8. [PMID: 40237797 DOI: 10.1007/s00210-025-04109-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025]
Abstract
Ulcerative colitis (UC) is one of the inflammatory bowel diseases characterized by colonic damage. Epigenetic mechanisms are suggested to play a role in the pathogenesis of UC. Pioglitazone has shown promise for the treatment of UC; however, the role of epigenetic pathways in this effect is unclear. The current study aimed to explore the therapeutic and protective effects of pioglitazone against acetic acid-induced colitis (AA-C) in rats and the role of epigenetic modulation and antioxidant mechanisms in this effect. Forty male albino rats were divided into four groups (n = 10/group): control (normal saline), acetic-acid-induced ulcerative colitis (AA-C) (3 days, 2 ml acetic acid 4%), pioglitazone-treated (AA, followed by 3-week oral pioglitazone 25 mg/kg/day), and pioglitazone-protected groups (3-day oral pioglitazone 25 mg/kg/day before AA, continued with AA, and 3 weeks later). After the experiment, the body weight, colon weight-to-length ratio, and colonic tissue were evaluated. The colonic expression of epigenetic markers (DNA methyltransferase- 1 and methylated E-cadherin), oxidative stress marker (malondialdehyde), antioxidant enzyme (superoxide dismutase), and angiotensin-converting enzyme- 2 (ACE- 2) was evaluated. The pioglitazone-protected and treated groups showed significant inhibition of DNA methyltransferase- 1 and methylated E-cadherin with improvement in colonic tissue macroscopic and microscopic signs of inflammation, improved weight, less oxidative stress, and less ACE- 2 expression. These beneficial actions were more pronounced among the pioglitazone-protected group. Pioglitazone could mitigate AA-C in rats by inhibiting epigenetic DNA methyltransferase- 1 and E-cadherin gene methylation. It also inhibits oxidative stress and prevents the overexpression of ACE- 2.
Collapse
Affiliation(s)
- Suzan Awad AbdelGhany Morsy
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Dr. Fahmi Abdelmeguid St., Mowassah Campus, Alexandria, 21561, Egypt.
- Pathological Sciences Department, MBBS Program, Fakeeh College for Medical Sciences, 21461, Jeddah, Saudi Arabia.
| | - Lobna M M A Abd El Mottelib
- Department of Human Anatomy and Embryology, Faculty of Medicine, Alexandria University, Dr. Fahmi Abdelmeguid St., Mowassah Campus, Alexandria, 21561, Egypt
| | - Sara Assem
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Dr. Fahmi Abdelmeguid St., Mowassah Campus, Alexandria, 21561, Egypt
| | - M M Abd El Aziz
- Department of Pathology, Faculty of Medicine, Alexandria University, Dr. Fahmi Abdelmeguid St., Mowassah Campus, Alexandria, 21561, Egypt
| | - Anne H Elgeziry
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Dr. Fahmi Abdelmeguid St., Mowassah Campus, Alexandria, 21561, Egypt
| |
Collapse
|
8
|
Li B, Li X, Tang X, Wang J. Prediction and Evaluation of Coronavirus and Human Protein-Protein Interactions Integrating Five Different Computational Methods. Proteins 2025. [PMID: 40231383 DOI: 10.1002/prot.26826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 03/08/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025]
Abstract
The high lethality and infectiousness of coronaviruses, particularly SARS-Cov-2, pose a significant threat to human society. Understanding coronaviruses, especially the interactions between these viruses and humans, is crucial for mitigating the coronavirus pandemic. In this study, we conducted a comprehensive comparison and evaluation of five prevalent computational methods: interolog mapping, domain-domain interaction methodology, domain-motif interaction methodology, structure-based approaches, and machine learning techniques. These methods were assessed using unbiased datasets that include C1, C2h, C2v, and C3 test sets. Ultimately, we integrated these five methodologies into a unified model for predicting protein-protein interactions (PPIs) between coronaviruses and human proteins. Our final model demonstrates relatively better performance, particularly with the C2v and C3 test sets, which are frequently used datasets in practical applications. Based on this model, we further established a high-confidence PPI network between coronaviruses and humans, consisting of 18,012 interactions between 3843 human proteins and 129 coronavirus proteins. The reliability of our predictions was further validated through the current knowledge framework and network analysis. This study is anticipated to enhance mechanistic understanding of the coronavirus-human relationship a while facilitating the rediscovery of antiviral drug targets. The source codes and datasets are accessible at https://github.com/covhppilab/CoVHPPI.
Collapse
Affiliation(s)
- Binghua Li
- Key Laboratory of Smart Farming for Agricultural Animals, Huazhong Agricultural University, Wuhan, China
- Hubei Key Laboratory of Agricultural Bioinformatics, Huazhong Agricultural University, Wuhan, China
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Xiaoyu Li
- Key Laboratory of Smart Farming for Agricultural Animals, Huazhong Agricultural University, Wuhan, China
- Hubei Key Laboratory of Agricultural Bioinformatics, Huazhong Agricultural University, Wuhan, China
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Xian Tang
- Key Laboratory of Smart Farming for Agricultural Animals, Huazhong Agricultural University, Wuhan, China
- Hubei Key Laboratory of Agricultural Bioinformatics, Huazhong Agricultural University, Wuhan, China
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Jia Wang
- Key Laboratory of Smart Farming for Agricultural Animals, Huazhong Agricultural University, Wuhan, China
- Hubei Key Laboratory of Agricultural Bioinformatics, Huazhong Agricultural University, Wuhan, China
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
9
|
Chen VHE, Tay JK, Gurung R, Nair S, Tay DJW, Tan KS, Foo RSY, Tambyah PA. ACE2 and TMPRSS2 gene expression is reduced acutely in SARS-CoV-2 patients but returns to normal with recovery. Sci Rep 2025; 15:12828. [PMID: 40229332 PMCID: PMC11997096 DOI: 10.1038/s41598-025-96279-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 03/27/2025] [Indexed: 04/16/2025] Open
Abstract
During the SARS-CoV-2 pandemic, angiotensin-converting enzyme 2 (ACE2) was identified as the major entry receptor for the virus and transmembrane serine protease 2 (TMPRSS2) as an important SARS-CoV-2 entry factor. Previous studies investigating the impact of ACE2 and TMPRSS2 gene expression on SARS-CoV-2 susceptibility in adults have yielded inconsistent results, thereby underscoring the need for further research in this domain. We obtained nasopharyngeal swabs from infected adults during the acute and late convalescent phase of SARS-CoV-2 infection and compared the expression of both genes with non-infected household member contacts. We found that ACE2 and TMPRSS2 gene expression is temporarily reduced during the acute phase of SARS-CoV-2 infection presumably due to viral disruption of transcription. Post-recovery, however, the expression of ACE2 and TMPRSS2 was comparable to non-infected household contacts. The lack of significant differences in ACE2 and TMPRSS2 gene expression between SARS-CoV-2-positive adults and uninfected household controls suggests that factors influencing susceptibility to SARS-CoV-2 infection in adults may extend beyond ACE2 and TMPRSS2 expression. However, the findings should be interpreted with caution due to the study's limited sample size and the heterogeneity of COVID-19 cases. Other non-physiological factors, such as enhanced hygiene practices following the infection of a household member, may also contribute to absence of infection among healthy controls.
Collapse
Affiliation(s)
- Vanessa Hui En Chen
- Department of Otolaryngology - Head and Neck Surgery, National University of Singapore, 1E Kent Ridge Road, 119228, Singapore, Singapore
| | - Joshua K Tay
- Department of Otolaryngology - Head and Neck Surgery, National University of Singapore, 1E Kent Ridge Road, 119228, Singapore, Singapore.
- Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Rijan Gurung
- Cardiovascular-Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Smeetha Nair
- Department of Medicine, National University of Singapore, 1E Kent Ridge Road, 119228, Singapore, Singapore
- Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Douglas Jie Wen Tay
- Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kai Sen Tan
- Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Roger S Y Foo
- Cardiovascular-Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Paul A Tambyah
- Department of Medicine, National University of Singapore, 1E Kent Ridge Road, 119228, Singapore, Singapore.
- Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
10
|
Yan C, Dong T, Shan Y, Zhao B, Yang H, Cai Y, Li S, Liu Q, Chu Y, Hao H, Cheng Z, Liu M, Zhang Y. Mycoplasma ovipnuemoniae impairs the immune response of sheep and suppresses neutrophil function by inhibiting S100A9. Vet Microbiol 2025; 303:110446. [PMID: 40022823 DOI: 10.1016/j.vetmic.2025.110446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Mycoplasma pneumonia is a chronic respiratory disease that seriously affects the health of sheep. To date, little information is available about the damage caused by Mycoplasma ovipneumoniae (MO) pneumonia to host lungs. Here, after sheep were infected with MO for 28 days, severe inflammatory reactions and pathological damage occurred. By using single-cell RNA sequencing (scRNA-seq), all the transcriptome changes in 11 cell types in sheep lung tissue were systematically analyzed, and the key biological processes regulating inflammation and immunity were identified. Moreover, we constructed both intercellular communication models and differential expression maps of key regulatory genes for each cell subgroup. We also specifically focused on the response of T cell subpopulations and neutrophils to MO infection. Long-term infection may affect an organism's immune response, inhibit intercellular communication, and highlight the important role of the cyclophilin A (CypA) and macrophage migration inhibitory factor (MIF) pathways in intercellular communication. Notably, MO infection decreased the toxicity of CD8 effector T cells and depleted regulatory T cells, thus inhibiting normal cell function. Subsequently, emphasis was placed on the important role of the neutrophil marker gene S100A9 in promoting neutrophil clearance of MO through activation of the ERK signaling pathway and reactive oxygen species (ROS) burst in vitro. These results contribute to understanding the progression of MO infection in the lungs and provide a rich database on the molecular basis of the response to different cell types in MO infection.
Collapse
Affiliation(s)
- Chenbo Yan
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Tianning Dong
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Yiyi Shan
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Bingru Zhao
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Hua Yang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Cai
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Shanglai Li
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Qiuyue Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuefeng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Huafang Hao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Zilong Cheng
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Maojun Liu
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China; Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Yanli Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
11
|
Du Y, Gao J, He M, Yi M, Wu J, Feng L, Zeng B, Li Y, He R, Wang Y, Qin CF, Cui Z, Wang C. Simultaneous Blockade of CD209 and CD209L by Monoclonal Antibody Does Not Provide Sufficient Protection Against Multiple Viral Infections In Vivo. Immunology 2025; 174:411-422. [PMID: 39783143 DOI: 10.1111/imm.13889] [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: 09/19/2024] [Revised: 11/18/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Many virus species, including Ebola virus, Marburg virus, SARS-CoV-2, dengue virus (DENV) and Zika virus (ZIKV), exploit CD209 and CD209L as alternative or attachment receptors for viral cis- or trans-infection. Thus, CD209 and CD209L may be critical targets for the development of therapeutic monoclonal blocking antibody drugs to disrupt the infection process caused by multiple viruses. Here, we produced a human chimeric monoclonal blocking antibody that simultaneously blocks CD209 and CD209L, namely 7-H7-B1. We show that 7-H7-B1 effectively blocks multiple pseudotyped or live viral infections in vitro, including SARS-CoV, SARS-CoV-2, Ebola virus, Marburg virus, ZIKV and DENV infections. However, the 7-H7-B1 mAb does not provide favourable protection against Zaire Ebola virus or ZIKV infection in hCD209 knock-in mice in vivo. Thus, our findings indicate that although CD209 and CD209L are critical for multiple viral infections in vitro, they may play only a partial role in viral infections in vivo.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Cell Adhesion Molecules/immunology
- Cell Adhesion Molecules/antagonists & inhibitors
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/immunology
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/antagonists & inhibitors
- Lectins, C-Type/immunology
- Lectins, C-Type/antagonists & inhibitors
- Zika Virus/immunology
- SARS-CoV-2/immunology
- Ebolavirus/immunology
- Zika Virus Infection/immunology
- Antibodies, Blocking/pharmacology
- Antibodies, Blocking/immunology
- Dengue Virus
- Vero Cells
- Virus Diseases/immunology
- Receptors, Virus/antagonists & inhibitors
- Receptors, Virus/immunology
Collapse
Affiliation(s)
- Yanyun Du
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jiawang Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, P.R. China
- University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Mengjiao He
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming Yi
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jiaqi Wu
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Lingyun Feng
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Bo Zeng
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yangyang Li
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ruirui He
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yuan Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, P.R. China
- University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Chenhui Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Jiang Z, Zhai C, Tang G. Novel Antihypertensive Medications to Target the Renin-Angiotensin System: Mechanisms and Research. Rev Cardiovasc Med 2025; 26:27963. [PMID: 40351692 PMCID: PMC12059749 DOI: 10.31083/rcm27963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/30/2024] [Accepted: 02/10/2025] [Indexed: 05/14/2025] Open
Abstract
An estimated 1.28 billion individuals in the global population suffer from hypertension. Importantly, uncontrolled hypertension is strongly linked to various cardiovascular and cerebrovascular diseases. The role of the renin-angiotensin system (RAS) is widely acknowledged in the development and progression of hypertension. This system comprises angiotensinogen, the renin/(pro)renin/(pro)renin receptor (PRR) axis, the renin/angiotensin-converting enzyme/angiotensin (Ang) II/Ang II type I receptor (AT1R) axis, the renin/angiotensin-converting enzyme (ACE) 2/Ang (1-7)/Mas receptor (MasR) axis, the alamandine/Mas-related G protein-coupled D (MrgD) receptor axis, and the renin/ACE/Ang II/Ang II type II receptor (AT2R) axis. Additionally, brain Ang III plays a vital role in regulating central blood pressure. The current overview presents the latest research findings on the mechanisms through which novel anti-hypertensive medications target the RAS. These include zilebesiran (targeting angiotensinogen), PRO20 (targeting the renin/(pro)renin/PRR axis), sacubitril/valsartan (targeting the renin/ACE/Ang II/AT1R axis), GSK2586881, Ang (1-7) and AVE0991 (targeting the renin/ACE2/Ang (1-7)/MasR axis), alamandine (targeting the alamandine/MrgD receptor axis), C21 and β-Pro7-Ang III (targeting the renin/ACE/Ang II/AT2R axis), EC33, and firibastat and NI956 (targeting brain Ang III).
Collapse
Affiliation(s)
- Zhe Jiang
- Department of Cardiology, Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Changlin Zhai
- Department of Cardiology, The Affiliated Hospital of Jiaxing University, 314001 Jiaxing, Zhejiang, China
| | - Guanmin Tang
- Department of Cardiology, The Affiliated Hospital of Jiaxing University, 314001 Jiaxing, Zhejiang, China
| |
Collapse
|
13
|
Hiti L, Markovič T, Lainscak M, Farkaš Lainščak J, Pal E, Mlinarič-Raščan I. The immunopathogenesis of a cytokine storm: The key mechanisms underlying severe COVID-19. Cytokine Growth Factor Rev 2025; 82:1-17. [PMID: 39884914 DOI: 10.1016/j.cytogfr.2024.12.003] [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: 10/22/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025]
Abstract
A cytokine storm is marked by excessive pro-inflammatory cytokine release, and has emerged as a key factor in severe COVID-19 cases - making it a critical therapeutic target. However, its pathophysiology was poorly understood, which hindered effective treatment. SARS-CoV-2 initially disrupts angiotensin signalling, promoting inflammation through ACE-2 downregulation. Some patients' immune systems then fail to shift from innate to adaptive immunity, suppressing interferon responses and leading to excessive pyroptosis and neutrophil activation. This amplifies tissue damage and inflammation, creating a pro-inflammatory loop. The result is the disruption of Th1/Th2 and Th17/Treg balances, lymphocyte exhaustion, and extensive blood clotting. Cytokine storm treatments include glucocorticoids to suppress the immune system, monoclonal antibodies to neutralize specific cytokines, and JAK inhibitors to block cytokine receptor signalling. However, the most effective treatment options for mitigating SARS-CoV-2 infection remain vaccines as a preventive measure and antiviral drugs for the early stages of infection. This article synthesizes insights into immune dysregulation in COVID-19, offering a framework to better understand cytokine storms and to improve monitoring, biomarker discovery, and treatment strategies for COVID-19 and other conditions involving cytokine storms.
Collapse
Affiliation(s)
- Luka Hiti
- Faculty of Pharmacy, University of Ljubljana, Slovenia
| | | | - Mitja Lainscak
- General Hospital Murska Sobota, Slovenia; Faculty of Medicine, University of Ljubljana, Slovenia
| | | | - Emil Pal
- General Hospital Murska Sobota, Slovenia
| | | |
Collapse
|
14
|
Brügger M, Machahua C, Zumkehr T, Cismaru C, Jandrasits D, Trüeb B, Ezzat S, Oliveira Esteves BI, Dorn P, Marti TM, Zimmer G, Thiel V, Funke-Chambour M, Alves MP. Aging shapes infection profiles of influenza A virus and SARS-CoV-2 in human precision-cut lung slices. Respir Res 2025; 26:112. [PMID: 40128814 PMCID: PMC11934781 DOI: 10.1186/s12931-025-03190-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) outbreak revealed the susceptibility of elderly patients to respiratory virus infections, showing cell senescence or subclinical persistent inflammatory profiles and favoring the development of severe pneumonia. METHODS In our study, we evaluated the potential influence of lung aging on the efficiency of replication of influenza A virus (IAV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as well as determining the pro-inflammatory and antiviral responses of the distal lung tissue. RESULTS Using precision-cut lung slices (PCLS) from donors of different ages, we found that pandemic H1N1 and avian H5N1 IAV replicated in the lung parenchyma with high efficacy. In contrast to these IAV strains, SARS-CoV-2 Early isolate and Delta variant of concern (VOC) replicated less efficiently in PCLS. Interestingly, both viruses showed reduced replication in PCLS from older compared to younger donors, suggesting that aged lung tissue represents a suboptimal environment for viral replication. Regardless of the age-dependent viral loads, PCLS responded to H5N1 IAV infection by an induction of IL-6 and IP10/CXCL10, both at the mRNA and protein levels, and to H1N1 IAV infection by induction of IP10/CXCL10 mRNA. Finally, while SARS-CoV-2 and H1N1 IAV infection were not causing detectable cell death, H5N1 IAV infection led to more cytotoxicity and induced significant early interferon responses. CONCLUSIONS In summary, our findings suggest that aged lung tissue might not favor viral dissemination, pointing to a determinant role of dysregulated immune mechanisms in the development of severe disease.
Collapse
Affiliation(s)
- Melanie Brügger
- Institute of Virology and Immunology, Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| | - Carlos Machahua
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Trix Zumkehr
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Christiana Cismaru
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Institute of Virology, Freie Universitaet Berlin, Berlin, Germany
| | - Damian Jandrasits
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Spiez Laboratory, Federal Office for Civil Protection, Spiez, Switzerland
| | - Bettina Trüeb
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Sara Ezzat
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Blandina I Oliveira Esteves
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Thomas M Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Gert Zimmer
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Volker Thiel
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases (MCID), University of Bern, Bern, Switzerland
- European Virus Bioinformatics Center, Jena, Germany
| | - Manuela Funke-Chambour
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marco P Alves
- Institute of Virology and Immunology, Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
- Multidisciplinary Center for Infectious Diseases (MCID), University of Bern, Bern, Switzerland.
| |
Collapse
|
15
|
Kameni MN, Tchoupe EB, Kamdem SD, Bhalla N, Assam Assam JP, Tepa AN, Neba FR, Nanda RK, Awuah AAA, Amuasi JH, Netongo PM. Mutations in ace2 gene modulate cytokine levels and alter immune responses in Mycobacterium tuberculosis and SARS-CoV-2 co-infection: a Cameroonian cohort. Front Immunol 2025; 16:1533213. [PMID: 40196114 PMCID: PMC11973369 DOI: 10.3389/fimmu.2025.1533213] [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: 11/23/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction SARS-CoV-2 and Mycobacterium tuberculosis (Mtb) share similarities in their modes of transmission, pathophysiological symptoms, and clinical manifestations. An imbalance in the immune response characterised by elevated levels of some inflammatory cytokines caused by tuberculosis (TB) and COVID-19 may increase the risk of developing a severe disease-like condition. It has been reported that TB increases the expression levels of Ace2 (angiotensin converting enzyme 2) and Tmprss2 (transmembrane protease serine 2) proteins, which are essential for COVID-19 pathogenesis. Single nucleotide polymorphisms (SNPs) variants of ace2 and tmprss2 genes can impact virus and host-cell interactions and alter immune responses by modulating cytokine production. This may modify the susceptibility and/or severity in COVID-19-infected people. The role of SNPs in ace2 and tmprss2 in relation to Mtb and SARS-CoV-2 co-infection is relatively underexplored. Method In this study, genotype frequency of 10 SNPs of ace2 and 03 SNPs of tmprss2 genes in a Cameroonian cohort consisting of COVID-19-positive (n = 31), TB-positive (n = 43), TB-COVID-19 co-infected (n = 21), and a control group (n = 24) were studied. The immune response was estimated by quantitating inflammatory cytokine levels alongside self-reported and clinically diagnosed symptoms. The relationship between specific genetic mutations in these ace2 gene SNPs and their impact on cytokine expression levels in Mtb and SARS-CoV-2 co-infected patients was investigated. Results We identified wild-type, heterozygous, and double-mutant genotypes in seven SNPs (rs2285666, rs6632677, rs4646116, rs4646140, rs147311723, rs2074192 and rs4646142) in ace2 gene, which showed significant variations in distribution across the study groups. Our most significant findings include the association of double mutant alleles (AA) of rs4646140 and rs2074192 in the ace2 gene with decreased IL-6 and IL-2 expression levels respectively in TB-COVID-19 participants. Also, the double mutant alleles (AA) of rs4646116 were responsible for increased expression level of IL-2 in TB-COVID-19 patients. Additionally, elevated serum levels of AST, urea, and D-dimer, as well as increased plasma concentrations of IL-10, IFN-γ, and TNF-α, have been associated with co-infections involving Mtb and SARS-CoV-2. Conclusion These biomarkers may reflect the complex interplay between the two pathogens and their impact on host immune responses and disease progression. This study highlights the critical role of genetic and immunological factors in shaping altered immune responses during co-infections involving Mtb and SARS-CoV-2. By elucidating these factors, the findings provide a foundation for a deeper understanding of host-pathogen interactions and their implications for disease progression and outcomes. Furthermore, this research has the potential to drive advancements in diagnostic approaches enabling more accurate detection and monitoring of co-infections.
Collapse
Affiliation(s)
- Mary Ngongang Kameni
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Department of Microbiology, University of Yaounde I, Yaounde, Cameroon
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Eric Berenger Tchoupe
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Department of Clinical Biochemistry, Faculty of Medicine and Biomedical Science, University of Yaounde I, Yaounde, Cameroon
| | - Severin Donald Kamdem
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Department of Pathology, School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Nikhil Bhalla
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | | | - Arnaud Njuiget Tepa
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
| | - Fuh Roger Neba
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
| | - Ranjan Kumar Nanda
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Anthony Afum-Adjei Awuah
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - John Humphrey Amuasi
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Palmer Masumbe Netongo
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Biology Program, School of Science, Navajo Technical University, Crownpoint, NM, United States
- Department of Biochemistry, University of Yaounde I, Yaounde, Cameroon
| |
Collapse
|
16
|
Tavelli A, Vergori A, Cingolani A, Bai F, Azzini AM, Hara GL, Caponcello MG, Rinaldi M, Palacios-Baena ZR, Gatti M, Maccarrone G, Tacconelli E, Antinori A, Monforte AD. ORCHESTRA Delphi consensus: clinical management of SARS-CoV-2 infection in people with HIV. Clin Microbiol Infect 2025:S1198-743X(25)00124-7. [PMID: 40122207 DOI: 10.1016/j.cmi.2025.03.006] [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: 12/19/2024] [Revised: 02/21/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVES The interaction between HIV and COVID-19 resulted in a syndemic that showed an excess burden of disease for people with HIV (PWH). Four years of the COVID-19 pandemic have raised many unsolved questions about the optimal care of COVID-19 in PWH. METHODS We performed a study using a three-round Delphi methodology involving a panel of physicians with expertise in HIV and COVID-19 infections. The main aim of the study was to provide recommendations on critical clinical issues of COVID-19 among PWH and to inform physicians and policy-makers for improving care and prevention of COVID-19 in PWH. A total of 27 questions were conceived, focusing on four main areas of interest in the management of COVID-19 in PWH; a panel of 34 experts in HIV and COVID-19 care expressed their level of agreement on each item. Questions that received agreement/disagreement ≥79.4% of panellists were identified and statements were generated accordingly. RESULTS Consensus was reached on 19/27 items, resulting in 18 final statements. These statements addressed: (a) risk of COVID-19 progression to severe disease among PWH; (b) COVID-19 diagnostics and laboratory procedures; (c) early treatments with antivirals and/or monoclonal antibodies; (d) use of corticosteroids; (e) COVID-19 preventive strategies. DISCUSSION This consensus's study guides infectious diseases physicians in making decisions regarding the care of PWH for COVID-19, where results from the scientific literature are limited or conflicting.
Collapse
Affiliation(s)
- Alessandro Tavelli
- Icona Foundation, Milan, Italy; National PhD Programme in One Health Approaches to Infectious Diseases and Life Science Research, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Alessandra Vergori
- Clinical Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Antonella Cingolani
- Infectious Diseases Unit, Medical and Surgical Sciences Department, Fondazione Policlinico A. Gemelli, IRCCS, Università Cattolica Sacro Cuore, Rome, Italy.
| | - Francesca Bai
- Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Anna Maria Azzini
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Gabriel Levy Hara
- Instituto Alberto Taquini de Investigación en Medicina Traslacional, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria Giulia Caponcello
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Seville, Spain; Departamento de Medicina, Universidad de Sevilla, Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Seville, Spain; CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Matteo Rinaldi
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Zaira R Palacios-Baena
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Seville, Spain; Departamento de Medicina, Universidad de Sevilla, Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Seville, Spain; CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Milo Gatti
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Gaia Maccarrone
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Evelina Tacconelli
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Andrea Antinori
- Clinical Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | | |
Collapse
|
17
|
Breugem TI, Riesebosch S, Zhang J, Mykytyn AZ, Krabbendam L, Groen N, Baptista Varela S, Schipper D, van den Doel PB, van Acker R, Stadhouders R, Lamers MM, Haagmans BL. Variable DPP4 expression in multiciliated cells of the human nasal epithelium as a determinant for MERS-CoV tropism. Proc Natl Acad Sci U S A 2025; 122:e2410630122. [PMID: 40048293 PMCID: PMC11929475 DOI: 10.1073/pnas.2410630122] [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: 05/29/2024] [Accepted: 12/21/2024] [Indexed: 03/25/2025] Open
Abstract
Transmissibility of respiratory viruses is a complex viral trait that is intricately linked to tropism. Several highly transmissible viruses, including severe acute respiratory syndrome coronavirus 2 and Influenza viruses, specifically target multiciliated cells in the upper respiratory tract to facilitate efficient human-to-human transmission. In contrast, the zoonotic Middle East respiratory syndrome coronavirus (MERS-CoV) generally transmits poorly between humans, which is largely attributed to the absence of its receptor dipeptidyl peptidase 4 (DPP4) in the upper respiratory tract. At the same time, MERS-CoV epidemiology is characterized by occasional superspreading events, suggesting that some individuals can disseminate this virus effectively. Here, we utilized well-differentiated human pulmonary and nasal airway organoid-derived cultures to further delineate the respiratory tropism of MERS-CoV. We find that MERS-CoV replicated to high titers in both pulmonary and nasal airway cultures. Using single-cell messenger-RNA sequencing, immunofluorescence, and immunohistochemistry, we show that MERS-CoV preferentially targeted multiciliated cells, leading to loss of ciliary coverage. MERS-CoV cellular tropism was dependent on the differentiation of the organoid-derived cultures, and replication efficiency varied considerably between donors. Similarly, variable and focal expression of DPP4 was revealed in human nose tissues. This study indicates that the upper respiratory tract tropism of MERS-CoV may vary between individuals due to differences in DPP4 expression, providing an explanation for the unpredictable transmission pattern of MERS-CoV.
Collapse
Affiliation(s)
- Tim I. Breugem
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Samra Riesebosch
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Jingshu Zhang
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Anna Z. Mykytyn
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Lisette Krabbendam
- Pulmonary Medicine Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Nathalie Groen
- Single Cell Discoveries, Utrecht3584 BW, The Netherlands
| | - Sivana Baptista Varela
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Debby Schipper
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Petra B. van den Doel
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Romy van Acker
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Ralph Stadhouders
- Pulmonary Medicine Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Mart M. Lamers
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore169857, Singapore
| | - Bart L. Haagmans
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| |
Collapse
|
18
|
Subbarayan K, Bieber H, Massa C, Rodríguez FAE, Hossain SMAA, Neuder L, Wahbi W, Salo T, Tretbar S, Al-Samadi A, Seliger B. Link of TMPRSS2 expression with tumor immunogenicity and response to immune checkpoint inhibitors in cancers. J Transl Med 2025; 23:294. [PMID: 40055791 PMCID: PMC11887338 DOI: 10.1186/s12967-025-06177-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND SARS-CoV-2 and other viruses rely on the protease function of the TMPRSS2 protein to invade host cells. Despite cancer patients often experience poorer outcomes following SARS-CoV-2 infection, the role of TMPRSS2 in different cancer types has not yet been analyzed in detail. Therefore, the aim of the study was to determine the expression, function and clinical relevance of TMPRSS2 in tumors. METHODS Publicly accessible RNA sequencing data from tumors, adjacent tissues and whole blood samples of COVID-19 patients as well as data from human tumor epithelial and endothelial cells infected with SARS-CoV-2 were analyzed for TMPRSS2 expression and correlated to the expression of immune-relevant genes and clinical parameters. In vitro models of cells transfected with TMPRSS2 (TMPRSS2high), siTMPRSS2 or mock controls (TMPRSS2low cells) were analyzed by qPCR, flow cytometry, ELISA and Western blot for the expression of immune response-relevant molecules. Co-cultures of TMPRSS2 model systems with blood peripheral mononuclear cells were employed to evaluate immune cell migration, cytotoxicity and cytokine release. RESULTS Higher expression levels of TMPRSS2 were found in blood from patients infected with SARS-CoV-2, while TMPRSS2 expression levels significantly varied between the tumor types analyzed. TMPRSS2high tumor cells exhibit increased activity of the interferon (IFN) signal pathway accompanied by an increased expression of class I human leukocyte antigens (HLA-I) and programmed cell death ligand 1 (PD-L1) elevated interleukin 6 (IL-6) secretion and reduced NK cell-mediated cytotoxicity compared to TMPRSS2low mock controls. Treatment with a Janus kinase (JAK) 2 inhibitor or TMPRSS2-specific siRNA decreased TMPRSS2 expression. Co-cultures of the in vitro TMPRSS2 models with peripheral blood mononuclear cells in the presence of the immune checkpoint inhibitor nivolumab resulted in a significantly increased migration and infiltration of immune cells towards TMPRSS2high cells and a reduced release of the innate immunity-related cytokines CCL2 and CCL3. CONCLUSIONS This study provides novel insights into the role of TMPRSS2 in various tumor systems and the impact of SARS-CoV-2 infection on the host immunogenicity via the activation of immune-relevant pathways. These findings were linked to the efficacy of immune checkpoint inhibitor therapy, offering a potential alternative strategy to mitigate the severity of COVID-19.
Collapse
Affiliation(s)
| | - Helena Bieber
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Chiara Massa
- Institute of Translational Immunology, Faculty of Health Sciences, Brandenburg Medical School "Theodor Fontane", Brandenburg an der Havel, Germany
| | - Felipe Adonis Escalona Rodríguez
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Center for Protein Studies, Faculty of Biology, University of Havana (UH), Havana, Cuba
- NanoCancer, Molecular Immunology Center (CIM), Havana, Cuba
| | - S M Al Amin Hossain
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Lisa Neuder
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Wafa Wahbi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, 90014, Finland
| | - Sandy Tretbar
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Institute of Dentistry, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Barbara Seliger
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
- Institute of Translational Immunology, Faculty of Health Sciences, Brandenburg Medical School "Theodor Fontane", Brandenburg an der Havel, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.
| |
Collapse
|
19
|
Andrade Barboza C, Gonçalves LM, Pereira E, Cruz RD, Andrade Louzada R, Boulina M, Almaça J. SARS-CoV-2 Spike S1 Subunit Triggers Pericyte and Microvascular Dysfunction in Human Pancreatic Islets. Diabetes 2025; 74:355-367. [PMID: 39715591 PMCID: PMC11842606 DOI: 10.2337/db24-0816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
The COVID-19 pandemic has profoundly affected human health; however, the mechanisms underlying its impact on metabolic and vascular systems remain incompletely understood. Clinical evidence suggests that SARS-CoV-2 directly disrupts vascular homeostasis, with perfusion abnormalities observed in various tissues. The pancreatic islet, a key endocrine miniorgan reliant on its microvasculature for optimal function, may be particularly vulnerable. Studies have proposed a link between SARS-CoV-2 infection and islet dysfunction, but the mechanisms remain unclear. Here, we investigated how SARS-CoV-2 spike S1 protein affects human islet microvascular function. Using confocal microscopy and living pancreas slices from organ donors without diabetes, we show that a SARS-CoV-2 spike S1 recombinant protein activates pericytes, key regulators of islet capillary diameter and β-cell function, and induces capillary constriction. These effects are driven by a loss of ACE2 from pericytes' plasma membrane, impairing ACE2 activity and increasing local angiotensin II levels. Our findings highlight islet pericyte dysfunction as a potential contributor to the diabetogenic effects of SARS-CoV-2 and offer new insights into the mechanisms linking COVID-19, vascular dysfunction, and diabetes. ARTICLE HIGHLIGHTS Different components of the renin-angiotensin system are expressed by vascular cells in human pancreatic islets. The islet microvasculature is responsive to vasoactive angiotensin peptides. This pancreatic renin-angiotensin system is targeted upon incubation with a SARS-CoV-2 spike recombinant protein. SARS-CoV-2 spike activates pericytes and constricts capillaries in human islets. Islet vascular dysfunction could contribute to dysglycemia in some patients with COVID-19.
Collapse
Affiliation(s)
- Catarina Andrade Barboza
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Luciana Mateus Gonçalves
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Elizabeth Pereira
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL
| | - Roxana Diaz Cruz
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Ruy Andrade Louzada
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Maria Boulina
- Diabetes Research Institute, University of Miami Health System, Miami, FL
| | - Joana Almaça
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Health System, Miami, FL
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
20
|
Fratta Pasini AM, Stranieri C, Di Leo EG, Bertolone L, Aparo A, Busti F, Castagna A, Vianello A, Chesini F, Friso S, Girelli D, Cominacini L. Identification of Early Biomarkers of Mortality in COVID-19 Hospitalized Patients: A LASSO-Based Cox and Logistic Approach. Viruses 2025; 17:359. [PMID: 40143288 PMCID: PMC11946718 DOI: 10.3390/v17030359] [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: 12/05/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
This study aimed to identify possible early biomarkers of mortality among clinical and biochemical parameters, iron metabolism parameters, and cytokines detected within 24 h from admission in hospitalized COVID-19 patients. We enrolled 80 hospitalized patients (40 survivors and 40 non-survivors) with COVID-19 pneumonia and acute respiratory failure. The median time from the onset of COVID-19 symptoms to hospital admission was lower in non-survivors than survivors (p < 0.05). Respiratory failure, expressed as the ratio of arterial oxygen partial pressure to the fraction of inspired oxygen (P/F), was more severe in non-survivors than survivors (p < 0.0001). Comorbidities were similar in both groups. Among biochemical parameters and cytokines, eGFR and interleukin (IL)-1β were found to be significantly lower (p < 0.05), while LDH, IL-10, and IL-8 were significantly higher in non-survivors than in survivors (p < 0.0005, p < 0.05 and p < 0.005, respectively). Among other parameters, LDH values distribution showed the most significant difference between study groups (p < 0.0001). LASSO feature selection combined with Cox proportional hazards and logistic regression models was applied to identify features distinguishing between survivors and non-survivors. Both approaches highlighted LDH as the strongest predictor, with IL-22 and creatinine emerging in the Cox model, while IL-10, eGFR, and creatinine were influential in the logistic model (AUC = 0.744 for Cox, 0.723 for logistic regression). In a similar manner, we applied linear regression for predicting LDH levels, identifying the P/F ratio as the top predictor, followed by IL-10 and eGFR (NRMSE = 0.128). Collectively, these findings underscore LDH's critical role in mortality prediction, with P/F and IL-10 as key determinants of LDH increases in this Italian COVID-19 cohort.
Collapse
Affiliation(s)
- Anna Maria Fratta Pasini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Chiara Stranieri
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Edoardo Giuseppe Di Leo
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Lorenzo Bertolone
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Antonino Aparo
- Interdepartmental Laboratory of Medical Research, Research Center LURM, University of Verona, 37134 Verona, Italy;
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Annalisa Castagna
- Department of Medicine, Section of Internal Medicine B, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy (S.F.)
| | - Alice Vianello
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Fabio Chesini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Simonetta Friso
- Department of Medicine, Section of Internal Medicine B, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy (S.F.)
| | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Luciano Cominacini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| |
Collapse
|
21
|
Quinn AE, Zhao L, Bell SD, Huq MH, Fang Y. Exploring Asthma as a Protective Factor in COVID-19 Outcomes. Int J Mol Sci 2025; 26:1678. [PMID: 40004141 PMCID: PMC11855143 DOI: 10.3390/ijms26041678] [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: 01/19/2025] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Asthma has long been associated with increased susceptibility to viral respiratory infections, leading to significant exacerbations and poorer clinical outcomes. Contrarily and interestingly, emerging data and research surrounding the COVID-19 pandemic have shown that patients with asthma infected with SARS-CoV-2 experienced decreased severity of disease, lower hospitalization rates, as well as decreased morbidity and mortality. Research has shown that eosinophils could enhance immune defense against viral infections, while inhaled corticosteroids can assist in controlling systematic inflammation. Moreover, reduced ACE-2 expression in individuals with asthma may restrict viral entry, and the Th2 immune response may offset the Th1 response typically observed in severe COVID-19 patients. These factors may help explain the favorable outcomes seen in asthmatic patients during the COVID-19 pandemic. This review highlights potential protective mechanisms seen in asthmatic patients, including eosinophilia, the use of inhaled corticosteroids, reduced ACE-2 expression, and a dominate Th2 immune response. Such a study will be helpful to better manage patients with asthma who have contracted COVID-19.
Collapse
Affiliation(s)
- Anthony E. Quinn
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Lei Zhao
- The Department of Respiratory Medicine, the 2nd People’s Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei 230002, China;
| | - Scott D. Bell
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Muhammad H. Huq
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
22
|
Lee C, Khan R, Mantsounga CS, Sharma S, Pierce J, Amelotte E, Butler CA, Farinha A, Parry C, Caballero O, Morrison JA, Uppuluri S, Whyte JJ, Kennedy JL, Zhang X, Choudhary G, Olson RM, Morrison AR. IL-1β-driven NF-κB transcription of ACE2 as a Mechanism of Macrophage Infection by SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.24.630260. [PMID: 39763770 PMCID: PMC11703209 DOI: 10.1101/2024.12.24.630260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Coronavirus disease 2019 (COVID-19), caused by infection with the enveloped RNA betacoronavirus, SARS-CoV-2, led to a global pandemic involving over 7 million deaths. Macrophage inflammatory responses impact COVID-19 severity; however, it is unclear whether macrophages are infected by SARS-CoV-2. We sought to identify mechanisms regulating macrophage expression of ACE2, the primary receptor for SARS-CoV-2, and to determine if macrophages are susceptible to productive infection. We developed a humanized ACE2 (hACE2) mouse whereby hACE2 cDNA was cloned into the mouse ACE2 locus under control of the native promoter. We validated the susceptibility of hACE2 mice to SARS-CoV-2 infection relative to wild-type mice and an established K18-hACE2 model of acute fulminating disease. Intranasal exposure to SARS-CoV-2 led to pulmonary consolidations with cellular infiltrate, edema, and hemorrhage, consistent with pneumonia, yet unlike the K18-hACE2 model, hACE2 mice survived and maintained stable weight. Infected hACE2 mice also exhibited a unique plasma chemokine, cytokine, and growth factor inflammatory signature relative to K18-hACE2 mice. Infected hACE2 mice demonstrated evidence of viral replication in infiltrating lung macrophages, and infection of macrophages in vitro revealed a transcriptional profile indicative of altered RNA and ribosomal processing machinery as well as activated cellular antiviral defense. Macrophage IL-1β-driven NF-κB transcription of ACE2 was an important mechanism of dynamic ACE2 upregulation, promoting macrophage susceptibility to infection. Experimental models of COVID-19 that make use of native hACE2 expression will allow for mechanistic insight into factors that can either promote host resilience or increase susceptibility to worsening severity of infection.
Collapse
Affiliation(s)
- Cadence Lee
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Rachel Khan
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Chris S. Mantsounga
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Sheila Sharma
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Julia Pierce
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Elizabeth Amelotte
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Celia A. Butler
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Andrew Farinha
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Crystal Parry
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Olivya Caballero
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Jeremi A. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Saketh Uppuluri
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Jeffrey J. Whyte
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri Division of Research, Innovation and Impact, Columbia, Missouri, USA
| | - Joshua L. Kennedy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Arkansas Children’s Research Institute, Little Rock, Arkansas, USA
| | - Xuming Zhang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
- Cardiovascular Research Center, Lifespan Cardiovascular Research Institute, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Rachel M. Olson
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri Division of Research, Innovation and Impact, Columbia, Missouri, USA
| | - Alan R. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
- Lead contact and corresponding author
| |
Collapse
|
23
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [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: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
24
|
Adamopoulos PG, Bartzoka N, Tsiakanikas P, Scorilas A. Characterization of novel ACE2 mRNA transcripts: The potential role of alternative splicing in SARS-CoV-2 infection. Gene 2025; 936:149092. [PMID: 39549777 DOI: 10.1016/j.gene.2024.149092] [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: 08/20/2024] [Revised: 10/25/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
The human angiotensin converting enzyme 2 (ACE2) gene encodes a type I transmembrane protein, which is homologous to angiotensin I-converting enzyme (ACE) and belongs to the angiotensin-converting enzyme family of dipeptidyl carboxypeptidases. As highlighted by the COVID-19 pandemic, ACE2 is not only crucial for the renin-angiotensin-aldosterone system (RAAS), but also displays great affinity with the SARS-CoV-2 spike protein, representing the major receptor of the virus. Given the significance of ACE2 in COVID-19, especially among cancer patients, the present study aims to explore the transcriptional landscape of ACE2 in human cancer and non-cancerous cell lines through the design and implementation of a custom targeted long-read sequencing approach. Bioinformatics analysis of the massive parallel sequencing data led to the identification of novel ACE2 mRNA splice variants (ACE2 sv.7-sv.12) that demonstrate previously uncharacterized exon-skipping events as well as 5' and/or 3' alternative splice sites. Demultiplexing of the sequencing data elucidated the differential expression profile of the identified splice variants in multiple human cell types, whereas in silico analysis suggests that some of the novel splice variants could produce truncated ACE2 isoforms with altered functionalities, potentially influencing their interaction with the SARS-CoV-2 spike protein. In summary, our study sheds light on the complex alternative splicing landscape of the ACE2 gene in cancer cell lines, revealing novel splice variants that could have significant implications for SARS-CoV-2 susceptibility in cancer patients. These findings contribute to the increased understanding of ACE2's role in COVID-19 and highlight the importance of considering alternative splicing as a key factor in viral pathogenesis. Undoubtably, further research is needed to explore the functional roles of these variants and their potential as therapeutic targets in the ongoing fight against COVID-19.
Collapse
Affiliation(s)
- Panagiotis G Adamopoulos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Natalia Bartzoka
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Tsiakanikas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
25
|
Spiteri S, Salamon I, Girolamini L, Pascale MR, Marino F, Derelitto C, Caligaris L, Paghera S, Ferracin M, Cristino S. Surfaces environmental monitoring of SARS-CoV-2: Loop mediated isothermal amplification (LAMP) and droplet digital PCR (ddPCR) in comparison with standard Reverse-Transcription quantitative polymerase chain reaction (RT-qPCR) techniques. PLoS One 2025; 20:e0317228. [PMID: 39899502 PMCID: PMC11790120 DOI: 10.1371/journal.pone.0317228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/24/2024] [Indexed: 02/05/2025] Open
Abstract
The persistence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) on substrates, and the impact of fomites on Coronavirus Disease 19 (COVID-19) transmission, is until now, widely discussed. Consequently, further investigations are required for a correct risk assessment in high-risk facilities such as hospitals, healthcare facilities (HCFs), and long-term care facilities (LTCFs). Therefore, appropriate surveillance and disinfection programs represent the best approach to guarantee the safety of these communities. This study proposes an environmental SARS-CoV-2 surfaces routine monitoring approach in HCF and communities' settings, to provide rapid and effective evaluation of surface hygienic conditions and the effectiveness of applied sanitization measures. Surfaces samples (n = 118) were collected using the SRK® kit (Copan Italia) from 2020 to 2023. Three molecular techniques were compared: Reverse Transcription Loop mediated isothermal AMPlification (RT-LAMP, Enbiotech), Reverse-Transcription quantitative polymerase chain reaction (RT-qPCR) (RT-qPCR, Seegene) and droplet digital PCR (ddPCR, Bio-Rad). For ddPCR, two RNA extraction methods were compared: TRIzol LS (Invitrogen) versus QIAmp Viral Mini kit (QIAGEN), showing how the latter is more suitable for surfaces. Regarding the quantitative ddPCR results, the ROC analysis allowed to reduce the manufacturer cut-off for droplets number (from 3 to 1) for the positive samples. Moreover, a new cut-off for the viral RNA copies' number/μL for each target (N1 and N2) on environmental monitoring was fixed at 2,82. The results obtained using the QIAmp kit, suggested that the N2 target is more stable in the environment and could be most suitable for the virus environmental detection. The percentage of positive samples was similar among the techniques (26% for RT-LAMP, 36% for ddPCR and 23% for RT-qPCR). Using RT-qPCR as reference method, a sensitivity (SE) of 30% for RT-LAMP and 41% for ddPCR was observed. By contrast, specificity (SP) was higher for RT-LAMP (75%) respect to ddPCR (66%). Comparing the faster RT-LAMP with the sensitive ddPCR the 26% and 74% of SE and SP for RT-LAMP, were reported. The low sensitivity for RT-LAMP and ddPCR could be explained with the use of clinical rather than environmental kits, other than the changing in the virus prevalence during the sampling campaign. Although the RT-LAMP requires improvements in term of SE and SP, this research presents an innovative environmental monitoring and prevention method for SARS-CoV-2, that could be extended to other pathogens that are under environmental surveillance.
Collapse
Affiliation(s)
- Simona Spiteri
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Irene Salamon
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Luna Girolamini
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Maria Rosaria Pascale
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Federica Marino
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Carlo Derelitto
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Laura Caligaris
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | | | - Manuela Ferracin
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Sandra Cristino
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
26
|
Batlle D, Hassler L, Wysocki J. ACE2, From the Kidney to SARS-CoV-2: Donald Seldin Award Lecture 2023. Hypertension 2025; 82:166-180. [PMID: 39624896 DOI: 10.1161/hypertensionaha.124.22064] [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] [Indexed: 01/18/2025]
Abstract
ACE2 (angiotensin-converting enzyme 2) is a monocarboxypeptidase that cleaves Ang II (angiotensin II) among other substrates. ACE2 is present in the cell membrane of many organs, most abundantly in epithelial cells of kidney proximal tubules and the small intestine, and also exists in soluble forms in plasma and body fluids. Membrane-bound ACE2 exerts a renoprotective action by metabolizing Ang II and therefore attenuating the undesirable actions of excess Ang II. Therefore, soluble ACE2, by downregulating this peptide, may exert a therapeutic action. Our laboratory has designed ACE2 truncates that pass the glomerular filtration barrier to target the kidney renin-angiotensin system directly and, therefore, compensate for loss of kidney membrane-bound ACE2. Membrane-bound ACE2 is also the essential receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Soluble ACE2 proteins have been studied as a way to intercept SARS-CoV-2 from binding to membrane-bound ACE2 and prevent cell entry of SARS-CoV-2 altogether. We bioengineered a soluble ACE2 protein, termed ACE2 618-DDC-ABD, with increased binding affinity for SARS-CoV-2 and prolonged duration of action, which, when administered intranasally, provides near-complete protection from lethality in k18hACE2 mice infected with different SARS-CoV-2 variants. The main advantage of soluble ACE2 proteins for the neutralization of SARS-CoV-2 is their immediate onset of action and universality for current and future emerging SARS-CoV-2 variants. It is notable that ACE2 is critically involved in 2 dissimilar functions: as a receptor for cell entry of many coronaviruses and as an enzyme in the metabolism of Ang II, and yet in both cases, it is a therapeutic target.
Collapse
Affiliation(s)
- Daniel Batlle
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Luise Hassler
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jan Wysocki
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
27
|
Chang R, Chen SY, Hsieh TYJ, Chen HY, Wang SI, Hung YM, Wei JCC. Risk of SARS-CoV-2 infection and adverse outcomes among vaccinated patients with tuberculosis. Public Health 2025; 239:80-86. [PMID: 39798219 DOI: 10.1016/j.puhe.2024.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/04/2024] [Accepted: 09/30/2024] [Indexed: 01/15/2025]
Abstract
OBJECTIVES Limited data are available to assess breakthrough SARS-CoV-2 infections, medical utilization, and mortality in patients with tuberculosis (TB). The aim of this study was to examine the risk of COVID-19 and severe outcomes in patients with TB between January 2020 and March 2022. STUDY DESIGN US electronic medical records were used to identify TB and non-TB patients who completed the primary series of vaccination and had no prior COVID-19. METHODS Breakthrough infections and severe adverse outcomes, defined by the Centers for Disease Control and Prevention as hospitalization, need for mechanical ventilation and/or intensive care unit admission, or in-hospital mortality, following a positive SARS-CoV-2 polymerase chain reaction (PCR) test. Follow-up began 14 days after the primary vaccination series was completed and continued for 365 days. RESULTS The study included 15,541 TB and 15,541 non-TB patients. The risk of breakthrough infection was significantly higher in the TB group than in the non-TB group after controlling for age, sex, ethnicity, socioeconomic status, and lifestyles (adjusted HR, 1.444; 95 % CI, 1.321-1.579). Similar trends were observed in pre-specified subgroup analyses stratified by age, sex, and status of TB. Patients in the TB group had higher risks of emergency room visit and critical care admission [adjusted HRs, 1.244 (95 % CI, 1.175-1.316) and 1.404 (95 % CI, 1.182-1.668)]. CONCLUSIONS Our study revealed a higher risk of COVID-19 breakthrough infections and adverse outcomes among patients with TB. Thus, besides priority COVID-19 vaccination, healthcare providers should continue vigilance for patients with TB.
Collapse
Affiliation(s)
- Renin Chang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Recreation and Sports Management, Tajen University, Pintung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan.
| | - Sheng-Yin Chen
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA. USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Tina Yi-Jin Hsieh
- Department of Obstetrics & Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Obstetrics & Gynecology, Massachusetts General Hospital, Boston, MA, USA; Department of Bioinformatics, Harvard Medical School, Boston, MA, USA.
| | - Hui-Yuan Chen
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.
| | - Shiow-Ing Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Center for Health Data Science, Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Yao-Min Hung
- Department of Internal Medicine, Taitung Hospital, Ministry of Health and Welfare, Taitung, Taiwan; Master Program in Biomedicine, College of Science and Engineering, National Taitung University, Taitung, Taiwan; College of Health and Nursing, Meiho University, Pingtung, Taiwan.
| | - James Cheng-Chung Wei
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China; Department of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan; Institute of Medicine/Department of Nursing, Chung Shan Medical University, Taichung, Taiwan.
| |
Collapse
|
28
|
Tabatabaei FS, Shafeghat M, Azimi A, Akrami A, Rezaei N. Endosomal Toll-Like Receptors intermediate negative impacts of viral diseases, autoimmune diseases, and inflammatory immune responses on the cardiovascular system. Expert Rev Clin Immunol 2025; 21:195-207. [PMID: 39137281 DOI: 10.1080/1744666x.2024.2392815] [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: 04/30/2024] [Revised: 07/17/2024] [Accepted: 08/12/2024] [Indexed: 08/15/2024]
Abstract
INTRODUCTION Cardiovascular disease (CVD) is the leading cause of morbidity globally, with chronic inflammation as a key modifiable risk factor. Toll-like receptors (TLRs), pivotal components of the innate immune system, including TLR-3, -7, -8, and -9 within endosomes, trigger intracellular cascades, leading to inflammatory cytokine production by various cell types, contributing to systemic inflammation and atherosclerosis. Recent research highlights the role of endosomal TLRs in recognizing self-derived nucleic acids during sterile inflammation, implicated in autoimmune conditions like myocarditis. AREAS COVERED This review explores the impact of endosomal TLRs on viral infections, autoimmunity, and inflammatory responses, shedding light on their intricate involvement in cardiovascular health and disease by examining literature on TLR-mediated mechanisms and their roles in CVD pathophysiology. EXPERT OPINION Removal of endosomal TLRs mitigates myocardial damage and immune reactions, applicable in myocardial injury. Targeting TLRs with agonists enhances innate immunity against fatal viruses, lowering viral loads and mortality. Prophylactic TLR agonist administration upregulates TLRs, protecting against fatal viruses and improving survival. TLRs play a complex role in CVDs like atherosclerosis and myocarditis, with therapeutic potential in modulating TLR reactions for cardiovascular health.
Collapse
Affiliation(s)
- Fatemeh Sadat Tabatabaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Melika Shafeghat
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirali Azimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashley Akrami
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
29
|
Alirezaee A, Mirmoghtadaei M, Heydarlou H, Akbarian A, Alizadeh Z. Interferon therapy in alpha and Delta variants of SARS-CoV-2: The dichotomy between laboratory success and clinical realities. Cytokine 2025; 186:156829. [PMID: 39693873 DOI: 10.1016/j.cyto.2024.156829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024]
Abstract
The COVID-19 pandemic has caused significant morbidity and mortality worldwide. The emergence of the Alpha and Delta variants of SARS-CoV-2 has led to a renewed interest in using interferon therapy as a potential treatment option. Interferons are a group of signaling proteins produced by host cells in response to viral infections. They play a critical role in the innate immune response to viral infections by inducing an antiviral state in infected and neighboring cells. Interferon therapy has shown promise as a potential treatment option for COVID-19. In this review paper, we review the current knowledge regarding interferon therapy in the context of the Alpha and Delta variants of SARS-CoV-2 and discuss the challenges that must be overcome to translate laboratory findings into effective clinical treatments.
Collapse
Affiliation(s)
- Atefe Alirezaee
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Mirmoghtadaei
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanieh Heydarlou
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Asiye Akbarian
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Alizadeh
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
van Baarle D, Nawijn MC. Variant-specific local tissue response to SARS-CoV-2 in the nasal mucosa. Nat Immunol 2025; 26:152-154. [PMID: 39880999 DOI: 10.1038/s41590-024-02067-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Affiliation(s)
- Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Center for Infectious Diseases Prevention, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Martijn C Nawijn
- Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
31
|
Saha A, Ganguly A, Kumar A, Srivastava N, Pathak R. Harnessing Epigenetics: Innovative Approaches in Diagnosing and Combating Viral Acute Respiratory Infections. Pathogens 2025; 14:129. [PMID: 40005506 PMCID: PMC11858160 DOI: 10.3390/pathogens14020129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Acute respiratory infections (ARIs) caused by viruses such as SARS-CoV-2, influenza viruses, and respiratory syncytial virus (RSV), pose significant global health challenges, particularly for the elderly and immunocompromised individuals. Substantial evidence indicates that acute viral infections can manipulate the host's epigenome through mechanisms like DNA methylation and histone modifications as part of the immune response. These epigenetic alterations can persist beyond the acute phase, influencing long-term immunity and susceptibility to subsequent infections. Post-infection modulation of the host epigenome may help distinguish infected from uninfected individuals and predict disease severity. Understanding these interactions is crucial for developing effective treatments and preventive strategies for viral ARIs. This review highlights the critical role of epigenetic modifications following viral ARIs in regulating the host's innate immune defense mechanisms. We discuss the implications of these modifications for diagnosing, preventing, and treating viral infections, contributing to the advancement of precision medicine. Recent studies have identified specific epigenetic changes, such as hypermethylation of interferon-stimulated genes in severe COVID-19 cases, which could serve as biomarkers for early detection and disease progression. Additionally, epigenetic therapies, including inhibitors of DNA methyltransferases and histone deacetylases, show promise in modulating the immune response and improving patient outcomes. Overall, this review provides valuable insights into the epigenetic landscape of viral ARIs, extending beyond traditional genetic perspectives. These insights are essential for advancing diagnostic techniques and developing innovative treatments to address the growing threat of emerging viruses causing ARIs globally.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar 814152, India;
| | - Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, India;
| | - Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
32
|
Walsh JML, Miao VN, Owings AH, Tang Y, Bromley JD, Kazer SW, Kimler K, Asare C, Ziegler CGK, Ibrahim S, Jivanjee T, George M, Navia AW, Drake RS, Parker A, Billingsley BC, Dotherow P, Tarugu S, Kota SK, Laird H, Wichman TG, Davis YT, Dhaliwal NS, Pride Y, Guo Y, Senitko M, Harvey J, Bates JT, Diamond G, Garrett MR, Robinson DA, Frame IJ, Lyons JJ, Robinson TO, Shalek AK, Horwitz BH, Glover SC, Ordovas-Montanes J. Variants and vaccines impact nasal immunity over three waves of SARS-CoV-2. Nat Immunol 2025; 26:294-307. [PMID: 39833605 DOI: 10.1038/s41590-024-02052-z] [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: 04/16/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025]
Abstract
Viral variant and host vaccination status impact infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), yet how these factors shift cellular responses in the human nasal mucosa remains uncharacterized. We performed single-cell RNA sequencing (scRNA-seq) on nasopharyngeal swabs from vaccinated and unvaccinated adults with acute Delta and Omicron SARS-CoV-2 infections and integrated with data from acute infections with ancestral SARS-CoV-2. Patients with Delta and Omicron exhibited greater similarity in nasal cell composition driven by myeloid, T cell and SARS-CoV-2hi cell subsets, which was distinct from that of ancestral cases. Delta-infected samples had a marked increase in viral RNA, and a subset of PER2+EGR1+GDF15+ epithelial cells was enriched in SARS-CoV-2 RNA+ cells in all variants. Prior vaccination was associated with increased frequency and activation of nasal macrophages. Expression of interferon-stimulated genes negatively correlated with coronavirus disease 2019 (COVID-19) severity in patients with ancestral and Delta but not Omicron variants. Our study defines nasal cell responses and signatures of disease severity across SARS-CoV-2 variants and vaccination.
Collapse
Affiliation(s)
- Jaclyn M L Walsh
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vincent N Miao
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
| | - Anna H Owings
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ying Tang
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Joshua D Bromley
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Samuel W Kazer
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Kyle Kimler
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Chelsea Asare
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carly G K Ziegler
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
- Harvard Graduate Program in Biophysics, Cambridge, MA, USA
| | - Samira Ibrahim
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tasneem Jivanjee
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Micayla George
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew W Navia
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Riley S Drake
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam Parker
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Paul Dotherow
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Spurthi Tarugu
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sai K Kota
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Hannah Laird
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - T Grant Wichman
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yesenia T Davis
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Neha S Dhaliwal
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yilianys Pride
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yanglin Guo
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michal Senitko
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jessie Harvey
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - John T Bates
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gill Diamond
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Michael R Garrett
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - D Ashley Robinson
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - I J Frame
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jonathan J Lyons
- Division of Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA
| | - Tanya O Robinson
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alex K Shalek
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard Graduate Program in Biophysics, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bruce H Horwitz
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Sarah C Glover
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Medicine, Section of Gastroenterology and Hepatology, Tulane University, New Orleans, LA, USA
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA.
- Program in Immunology, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
33
|
Hara Y, Jha MK, Huang JY, Han Y, Langohr IM, Gaglia G, Zhu C, Piepenhagen P, Gayvert K, Lim WK, Asrat S, Nash S, Jacob‐Nara JA, Orengo JM, Bangari DS, de Rinaldis E, Mattoo H, Hicks A. The IL-4-IL-4Rα axis modulates olfactory neuroimmune signaling to induce loss of smell. Allergy 2025; 80:440-461. [PMID: 39418114 PMCID: PMC11804309 DOI: 10.1111/all.16338] [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: 06/14/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 10/19/2024]
Abstract
IL-4 and IL-13 have non-redundant effects in olfaction, with loss of smell in mice evoked only by intranasal administration of IL-4, but not IL-13. IL-4-evoked pathophysiological effects on olfaction is independent of compromised structural integrity of the olfactory neuroepithelium. IL-4-IL-4Rα signaling modulates neuronal crosstalk with immune cells, suggesting a functional link between olfactory impairment and neuroinflammation. Abbreviations: IL, interleukin; KO, knock-out; wk, week; WT, wild-type.
Collapse
Affiliation(s)
- Yannis Hara
- Type 2 Inflammation, Immunology and Inflammation, SanofiCambridgeMassachusettsUSA
| | - Mithilesh Kumar Jha
- Type 2 Inflammation, Immunology and Inflammation, SanofiCambridgeMassachusettsUSA
| | - Jeremy Y. Huang
- Precision Medicine and Computational Biology, SanofiCambridgeMassachusettsUSA
| | - Yingnan Han
- Precision Medicine and Computational Biology, SanofiCambridgeMassachusettsUSA
| | | | - Giorgio Gaglia
- Precision Medicine and Computational Biology, SanofiCambridgeMassachusettsUSA
| | - Cheng Zhu
- Precision Medicine and Computational Biology, SanofiCambridgeMassachusettsUSA
| | | | - Kaitlyn Gayvert
- Molecular Profiling and Data Science, Regeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Wei Keat Lim
- Molecular Profiling and Data Science, Regeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Seblewongel Asrat
- Immunology and Inflammation, Regeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Scott Nash
- Medical Affairs, Regeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | | | - Jamie M. Orengo
- Immunology and Inflammation, Regeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | | | | | - Hamid Mattoo
- Precision Medicine and Computational Biology, SanofiCambridgeMassachusettsUSA
| | - Alexandra Hicks
- Type 2 Inflammation, Immunology and Inflammation, SanofiCambridgeMassachusettsUSA
| |
Collapse
|
34
|
Eldien HMS, Almaeen AH, El Fath AA, Taha AE, Ahmed R, Elfadil H, Hetta HF. Unlocking the Potential of RNA Sequencing in COVID-19: Toward Accurate Diagnosis and Personalized Medicine. Diagnostics (Basel) 2025; 15:229. [PMID: 39857114 PMCID: PMC11763845 DOI: 10.3390/diagnostics15020229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/08/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
COVID-19 has caused widespread morbidity and mortality, with its effects extending to multiple organ systems. Despite known risk factors for severe disease, including advanced age and underlying comorbidities, patient outcomes can vary significantly. This variability complicates efforts to predict disease progression and tailor treatment strategies. While diagnostic and therapeutic approaches are still under debate, RNA sequencing (RNAseq) has emerged as a promising tool to provide deeper insights into the pathophysiology of COVID-19 and guide personalized treatment. A comprehensive literature review was conducted using PubMed, Scopus, Web of Science, and Google Scholar. We employed Medical Subject Headings (MeSH) terms and relevant keywords to identify studies that explored the role of RNAseq in COVID-19 diagnostics, prognostics, and therapeutics. RNAseq has proven instrumental in identifying molecular biomarkers associated with disease severity in patients with COVID-19. It allows for the differentiation between asymptomatic and symptomatic individuals and sheds light on the immune response mechanisms that contribute to disease progression. In critically ill patients, RNAseq has been crucial for identifying key genes that may predict patient outcomes, guiding therapeutic decisions, and assessing the long-term effects of the virus. Additionally, RNAseq has helped in understanding the persistence of viral RNA after recovery, offering new insights into the management of post-acute sequelae, including long COVID. RNA sequencing significantly improves COVID-19 management, particularly for critically ill patients, by enhancing diagnostic accuracy, personalizing treatment, and predicting therapeutic responses. It refines patient stratification, improving outcomes, and holds promise for targeted interventions in both acute and long COVID.
Collapse
Affiliation(s)
- Heba M. Saad Eldien
- Department of Anatomy, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
| | - Abdulrahman H. Almaeen
- Department of Pathology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Ahmed Abo El Fath
- Tropical Medicine and Gastroenterology Department, Assiut University Hospital, Assiut 71515, Egypt;
| | - Ahmed E. Taha
- Microbiology and Immunology Unit, Department of Pathology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia;
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Rehab Ahmed
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| | - Hassabelrasoul Elfadil
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| | - Helal F. Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| |
Collapse
|
35
|
Chau LF, Lillico S, Opriessnig T, Blake R, Tardy L, Lee CH, Maxwell S, Warren C, Thornton E, Mclaughlin CL, McLachlan G, Tait-Burkard C, Fletcher S, Anderson S, Brown S, Gibbard L, Tzelos T, MacMillan-Christensen D, Baillie JK, Dorward DA, Griffiths DJ, Grey F. Human ACE2 transgenic pigs are susceptible to SARS-CoV-2 and develop COVID-19-like disease. Nat Commun 2025; 16:766. [PMID: 39824810 PMCID: PMC11742018 DOI: 10.1038/s41467-024-54615-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 11/18/2024] [Indexed: 01/20/2025] Open
Abstract
Animal models that accurately reflect COVID-19 are vital for understanding mechanisms of disease and advancing development of improved vaccines and therapeutics. Pigs are increasingly recognized as valuable models for human disease due to their genetic, anatomical, physiological, and immunological similarities to humans, and they present a more ethically viable alternative to non-human primates. However, pigs are not susceptible to SARS-CoV-2 infection which limits their utility as a model. To address this, we have developed transgenic pigs expressing human ACE2 that are susceptible to SARS-CoV-2 infection. Following challenge, clinical signs consistent with COVID-19, including fever, coughing and respiratory distress were observed, with virus replication detected in the nasal turbinates, trachea and lungs up to the study endpoint, seven days post-infection. Notably, examination of tissues revealed immunopathology in the lungs consistent with histological changes observed in fatal human COVID-19 cases. This study establishes human ACE2 transgenic pigs as a large animal model that accurately reflects many aspects of COVID-19 disease.
Collapse
Affiliation(s)
- Long Fung Chau
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Simon Lillico
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | | | - Rosemary Blake
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Luc Tardy
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Chen-Hsuin Lee
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Scott Maxwell
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Claire Warren
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Elizabeth Thornton
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Catherine L Mclaughlin
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Gerry McLachlan
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Christine Tait-Burkard
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Sarah Fletcher
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | | - J Kenneth Baillie
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
- Baillie Gifford Pandemic Science Hub, University of Edinburgh, Edinburgh, UK
| | - David A Dorward
- Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh, UK
- Department of Pathology, Royal Infirmary, Edinburgh, UK
| | | | - Finn Grey
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
36
|
Zhou L, Zhang L, Xu F. The combined manifestations of dramatically sore throat, congested and edematous mucosa, no-swelling tonsil are specific in acute Omicron pharyngitis. BMC Infect Dis 2025; 25:29. [PMID: 39762748 PMCID: PMC11702273 DOI: 10.1186/s12879-024-10364-6] [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: 04/16/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE To identify specific clinical signs of Omicron pharyngitis infection. METHODS A clinical cross-sectional retrospective study was designed to analyze the primary symptoms of pharyngitis in outpatients seeking treatment for sore throat. Pharyngeal congestion, mucosal edema, were measured using a visual analogue assessment score (0-10) while the presence of ulcers, no-tonsil-swelling, no-tonsil-exudate. They were recorded as "yes" or "no "as two-Categorical data by two senior clinicians, respectively. Significant clinical signs were selected and combined to form a diagnostic panel using SPSS software to differentiate between Omicron pharyngitis and other sore throat cases. The efficiency of the panel was calculated. RESULTS A total of 39 sore throat patients were included in the study, including 15 confirmed cases of Omicron pharyngitis through nuclear acid or Sars-Cov-2 virus antigen testing, and 24 cases of common pharyngitis caused by other pathogens. Mucosal congestion and edema were identified as the most significant symptoms and consolidated into a single working group. When combined with the third significant symptom of no-tonsil-swelling, the three-sign-combined diagnostic panel was found to have a high diagnostic efficiency. Mucosal congestion and edema were the most significant signs. When mucosal congestion and edema were consolidated into a single working panel, the cut-off values were determined to be 7.5 and 1, respectively. When combined with the third significant symptom no-tonsil-swelling, the three-sign diagnostic panel was found to have a high diagnostic efficiency. When compared with the gold standard measurement of Sars-Cov-2 virus antigen or nucleic acid, the diagnostic panel has a sensitivity of 66.7% and a specificity of 91.7%. CONCLUSION A combination of three signs may be a useful diagnostic tool for Omicron pharyngitis. Clinical signs of dramatic mucosal congestion and edema, non-swollen tonsils are the characteristics of Omicron pharyngitis.
Collapse
Affiliation(s)
- Lei Zhou
- Department of Otolaryngology-Head and Neck Surgery, Zhongshan Hospital, Fudan University, Fenglin Road 180, Shanghai, 200032, China
| | - Lineng Zhang
- Department of Biotechnology and Biomedical Laboratory Sciences, Shanghai University of Medicine & Health Sciences Medical Technology College, Zhouzhu Road 279, Shanghai, 201318, China.
| | - Feng Xu
- Department of Otolaryngology-Head and Neck Surgery, Zhongshan Hospital, Fudan University, Fenglin Road 180, Shanghai, 200032, China.
| |
Collapse
|
37
|
Spalinger MR, Sanati G, Chatterjee P, Hai R, Li J, Santos AN, Nordgren TM, Tremblay ML, Eckmann L, Hanson E, Scharl M, Wu X, Boland BS, McCole DF. Tofacitinib Mitigates the Increased SARS-CoV-2 Infection Susceptibility Caused by an IBD Risk Variant in the PTPN2 Gene. Cell Mol Gastroenterol Hepatol 2025; 19:101447. [PMID: 39756517 PMCID: PMC11953972 DOI: 10.1016/j.jcmgh.2024.101447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND & AIMS Coronavirus disease (COVID-19), caused by severe acquired respiratory syndrome-Coronavirus-2 (SARS-CoV-2), triggered a global pandemic with severe medical and socioeconomic consequences. Although fatality rates are higher among the elderly and those with underlying comorbidities, host factors that promote susceptibility to SARS-CoV-2 infection and severe disease are poorly understood. Although individuals with certain autoimmune/inflammatory disorders show increased susceptibility to viral infections, there is incomplete knowledge of SARS-CoV-2 susceptibility in these diseases. The aim of our study was to investigate whether the autoimmunity risk gene, PTPN2, which also confers elevated risk to develop inflammatory bowel disease, affects susceptibility to SARS-CoV-2 viral uptake. METHODS Using samples from PTPN2 genotyped patients with inflammatory bowel disease, PTPN2-deficient mice, and human intestinal and lung epithelial cell lines, we investigated how PTPN2 affects expression of the SARS-CoV-2 receptor angiotensin converting enzyme 2 (ACE2), and uptake of virus-like particles expressing the SARS-CoV2 spike protein and live SARS-CoV-2 virus. RESULTS We report that the autoimmune PTPN2 loss-of-function risk variant rs1893217 promotes expression of the SARS-CoV-2 receptor, ACE2, and increases cellular entry of SARS-CoV-2 spike protein and live virus. Elevated ACE2 expression and viral entry were mediated by increased Janus kinase-signal transducers and activators of transcription signaling and were reversed by the Janus kinase inhibitor, tofacitinib. CONCLUSION Collectively, our findings uncover a novel risk biomarker for increased expression of the SARS-CoV-2 receptor and viral entry, and identify a clinically approved therapeutic agent to mitigate this risk.
Collapse
Affiliation(s)
- Marianne R Spalinger
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California; Department of Gastroenterology and Hepatology, University Hospital Zurich, and University of Zurich, Zurich, Switzerland
| | - Golshid Sanati
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Pritha Chatterjee
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Rong Hai
- Department of Microbiology and Plant Pathology, University of California Riverside, Riverside, California
| | - Jiang Li
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Alina N Santos
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Tara M Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California; Current position: College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| | - Michel L Tremblay
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Lars Eckmann
- Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Elaine Hanson
- Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, and University of Zurich, Zurich, Switzerland
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Monrovia, California
| | - Brigid S Boland
- Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Declan F McCole
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California.
| |
Collapse
|
38
|
Edwards CT, Karunakaran KA, Garcia E, Beutler N, Gagne M, Golden N, Aoued H, Pellegrini KL, Burnett MR, Honeycutt CC, Lapp SA, Ton T, Lin MC, Metz A, Bombin A, Goff K, Scheuermann SE, Wilkes A, Wood JS, Ehnert S, Weissman S, Curran EH, Roy M, Dessasau E, Paiardini M, Upadhyay AA, Moore IN, Maness NJ, Douek DC, Piantadosi A, Andrabi R, Rogers TR, Burton DR, Bosinger SE. Passive infusion of an S2-Stem broadly neutralizing antibody protects against SARS-CoV-2 infection and lower airway inflammation in rhesus macaques. PLoS Pathog 2025; 21:e1012456. [PMID: 39847599 PMCID: PMC11793774 DOI: 10.1371/journal.ppat.1012456] [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: 07/31/2024] [Revised: 02/04/2025] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
The continued evolution of SARS-CoV-2 variants capable of subverting vaccine and infection-induced immunity suggests the advantage of a broadly protective vaccine against betacoronaviruses (β-CoVs). Recent studies have isolated monoclonal antibodies (mAbs) from SARS-CoV-2 recovered-vaccinated donors capable of neutralizing many variants of SARS-CoV-2 and other β-CoVs. Many of these mAbs target the conserved S2 stem region of the SARS-CoV-2 spike protein, rather than the receptor binding domain contained within S1 primarily targeted by current SARS-CoV-2 vaccines. One of these S2-directed mAbs, CC40.8, has demonstrated protective efficacy in small animal models against SARS-CoV-2 challenge. As the next step in the pre-clinical testing of S2-directed antibodies as a strategy to protect from SARS-CoV-2 infection, we evaluated the in vivo efficacy of CC40.8 in a clinically relevant non-human primate model by conducting passive antibody transfer to rhesus macaques (RM) followed by SARS-CoV-2 challenge. CC40.8 mAb was intravenously infused at 10mg/kg, 1mg/kg, or 0.1 mg/kg into groups (n = 6) of RM, alongside one group that received a control antibody (PGT121). Viral loads in the lower airway were significantly reduced in animals receiving higher doses of CC40.8. We observed a significant reduction in inflammatory cytokines and macrophages within the lower airway of animals infused with 10mg/kg and 1mg/kg doses of CC40.8. Viral genome sequencing demonstrated a lack of escape mutations in the CC40.8 epitope. Collectively, these data demonstrate the protective efficiency of broadly neutralizing S2-targeting antibodies against SARS-CoV-2 infection within the lower airway while providing critical preclinical work necessary for the development of pan-β-CoV vaccines.
Collapse
Affiliation(s)
- Christopher T. Edwards
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Kirti A. Karunakaran
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States of America
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Matthew Gagne
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Hadj Aoued
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Kathryn L. Pellegrini
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Matthew R. Burnett
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Christopher Cole Honeycutt
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stacey A. Lapp
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Thang Ton
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Mark C. Lin
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Amanda Metz
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Andrei Bombin
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Sarah E. Scheuermann
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Amelia Wilkes
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Jennifer S. Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stephanie Ehnert
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stacey Weissman
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Elizabeth H. Curran
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Melissa Roy
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Evan Dessasau
- Division of Histology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Amit A. Upadhyay
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Ian N. Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Nicholas J. Maness
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Daniel C. Douek
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anne Piantadosi
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, California, United States of America
| | - Thomas R. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, California, United States of America
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
39
|
Nwabufo CK. Uncovering the impact of COVID-19-mediated bidirectional dysregulation of cytochrome P450 3A4 on systemic and pulmonary drug concentrations using physiologically based pharmacokinetic modeling. Drug Metab Dispos 2025; 53:100008. [PMID: 39884806 DOI: 10.1124/dmd.124.001893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 01/22/2025] Open
Abstract
Several clinical studies have shown that COVID-19 increases the systemic concentration of drugs in hospitalized patients with COVID-19. However, it is unclear how COVID-19-mediated bidirectional dysregulation of hepatic and pulmonary cytochrome P450 (CYP) 3A4 affects drug concentrations, especially in the lung tissue, which is most affected by the disease. Herein, physiologically based pharmacokinetic modeling was used to demonstrate the differences in systemic and pulmonary concentrations of 4 respiratory infectious disease drugs when CYP3A4 is concurrently downregulated in the liver and upregulated in the lung based on existing clinical data on COVID-19-CYP3A4 interactions at varying severity levels including outpatients, non-intensive care unit (ICU), and ICU patients. The study showed that hepatic metabolism is the primary determinant of both systemic and pulmonary drug concentrations despite the concurrent bidirectional dysregulation of liver and lung CYP3A4. ICU patients had the most systemic and pulmonary drug exposure, with a percentage increase in the area under the concentration-time curve in the plasma compartment of approximately 44%, 56%, 114%, and 196% for clarithromycin, nirmatrelvir, dexamethasone, and itraconazole, respectively, relative to the healthy group. Within the ICU cohort, clarithromycin exhibited its highest exposure in lung tissue mass with a fold change of 1189, whereas nirmatrelvir and dexamethasone showed their highest exposure in the plasma compartment, with fold changes of about 126 and 5, respectively, compared with the maximum therapeutic concentrations for their target pathogens. Itraconazole was significantly underexposed in the lung fluid compartment, potentially explaining its limited efficacy for the treatment of COVID-19. These findings underscore the importance of optimizing dosing regimens in at risk ICU patients to enhance both efficacy and safety profiles. SIGNIFICANCE STATEMENT: This study investigated whether COVID-19-mediated concurrent hepatic downregulation and pulmonary upregulation of cytochrome P450 (CYP) 3A4 leads to differences in the systemic and pulmonary concentrations of 4 respiratory medicines. The study demonstrated that intercompartmental differences in drug concentrations were driven by only hepatic CYP3A4 expression. This work suggests that ICU patients with significant COVID-19-CYP3A4 interactions may be at risk of clinically relevant COVID-19-drug interactions, highlighting the need for optimizing dosing regimens in this patient group to improve safety and efficacy.
Collapse
Affiliation(s)
- Chukwunonso K Nwabufo
- Current affiliation: Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Current affiliation: OneDrug Inc., Toronto, Ontario, Canada; Program in Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada; Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
40
|
Karavaizoglu C, Suleyman A, Dolu KO, Yucel E, Demirkale ZH, Ozdemir C, Tamay ZU. Different Aspects of COVID-19: "Stay at Home" Increased Cat and Mite Sensitivity in Preschool Children. Pediatr Pulmonol 2025; 60:e27422. [PMID: 39607346 DOI: 10.1002/ppul.27422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Indoor allergen exposure and senitization is expected to increase significantly during the COVID-19 pandemic. The aim of this study was to assess the impact of this altered allergen exposure on allergen sensitivity patterns in children with asthma. METHODS Demographic data, clinical characteristics, and laboratory findings of asthmatic children aged 5 years and younger were assessed May 2019 to May 2020, 1 year after the start of lockdown measures. Those asthmatic children were compared with age- and sex-matched asthmatic peers assessed before the pandemic. Group I included asthmatic children who underwent skin testing within 12 months of the start of the lockdown (n = 112), while Group II included asthmatic children whose skin testing was done before the lockdown (n = 224). RESULTS A total of 336 preschool children diagnosed with asthma were evaluated. House dust mite (HDM) and animal dander (cat, dog) senitization rates were significantly higher in Group I than in Group II (66.1% vs. 42.9%; OR: 2.5, 95% CI: 1.6-4.1, p < 0.001 and 8.0% vs. 1.3%; OR: 6.4, CI: 1.7-24.2, p = 0.006, respectively). However, the number of asthma attacks requiring systemic corticosteroid treatment was higher in Group I than in Group II (p = 0.001). The duration of lockdown emerged as a significant risk factor for HDM senitization (p < 0.001, OR: 2.6, 95% CI: 1.6-4.1) and animal allergen senitization (p = 0.006, OR: 6.4, 95% CI: 1.7-27.3). CONCLUSION During the pandemic, senitization to indoor allergens, including HDM and animal dander, increased significantly in asthmatic children. This may be attributed to changes in lifestyle, increased time spent indoors and increased pet ownership.
Collapse
Affiliation(s)
- Cagla Karavaizoglu
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ayse Suleyman
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Kazım Okan Dolu
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Esra Yucel
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Zeynep Hızlı Demirkale
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Cevdet Ozdemir
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
- Department of Pediatric Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey
| | - Zeynep Ulker Tamay
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
41
|
Wu Y, Cui Y, Zheng X, Yao X, Sun G. Integrated machine learning to predict the prognosis of lung adenocarcinoma patients based on SARS-COV-2 and lung adenocarcinoma crosstalk genes. Cancer Sci 2025; 116:95-111. [PMID: 39489517 PMCID: PMC11711064 DOI: 10.1111/cas.16384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024] Open
Abstract
Viruses are widely recognized to be intricately associated with both solid and hematological malignancies in humans. The primary goal of this research is to elucidate the interplay of genes between SARS-CoV-2 infection and lung adenocarcinoma (LUAD), with a preliminary investigation into their clinical significance and underlying molecular mechanisms. Transcriptome data for SARS-CoV-2 infection and LUAD were sourced from public databases. Differentially expressed genes (DEGs) associated with SARS-CoV-2 infection were identified and subsequently overlapped with TCGA-LUAD DEGs to discern the crosstalk genes (CGs). In addition, CGs pertaining to both diseases were further refined using LUAD TCGA and GEO datasets. Univariate Cox regression was conducted to identify genes associated with LUAD prognosis, and these genes were subsequently incorporated into the construction of a prognosis signature using 10 different machine learning algorithms. Additional investigations, including tumor mutation burden assessment, TME landscape, immunotherapy response assessment, as well as analysis of sensitivity to antitumor drugs, were also undertaken. We discovered the risk stratification based on the prognostic signature revealed that the low-risk group demonstrated superior clinical outcomes (p < 0.001). Gene set enrichment analysis results predominantly exhibited enrichment in pathways related to cell cycle. Our analyses also indicated that the low-risk group displayed elevated levels of infiltration by immunocytes (p < 0.001) and superior immunotherapy response (p < 0.001). In our study, we reveal a close association between CGs and the immune microenvironment of LUAD. This provides preliminary insight for further exploring the mechanism and interaction between the two diseases.
Collapse
Affiliation(s)
- Yanan Wu
- School of Public HealthNorth China University of Science and TechnologyTangshanChina
| | - Yishuang Cui
- School of Public HealthNorth China University of Science and TechnologyTangshanChina
| | - Xuan Zheng
- School of Public HealthNorth China University of Science and TechnologyTangshanChina
| | - Xuemin Yao
- School of Public HealthNorth China University of Science and TechnologyTangshanChina
| | - Guogui Sun
- School of Public HealthNorth China University of Science and TechnologyTangshanChina
| |
Collapse
|
42
|
Yu Y, Zhou G, Du J, Zhu H, Guan H, Bi Y, Zhang D. Hypophysitis after COVID-19 vaccination in a patient with Rathke's cleft cyst: A case report. Hum Vaccin Immunother 2024; 20:2297455. [PMID: 38174857 PMCID: PMC10773625 DOI: 10.1080/21645515.2023.2297455] [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: 10/12/2023] [Accepted: 12/17/2023] [Indexed: 01/05/2024] Open
Abstract
With the widespread vaccination of COVID-19 vaccine, a few cases have been reported that COVID-19 vaccine may cause endocrine disorders. A 59-y-old man presented with a loss of appetite after the first COVID-19 vaccination, which resolved spontaneously after 3 d. After the second COVID-19 vaccination, the symptoms including the loss of appetite, nausea, and vomiting reappeared and worsened along with loss of vision. He was found to have severe hyponatremia, and further investigations revealed secondary adrenal insufficiency, secondary hypothyroidism and Rathke's cleft cyst. The patient responded well to glucocorticoid and levothyroxine supplementation, and at 1-y follow-up the patient developed hypogonadism. We hypothesize that hypophysitis is probably induced by COVID-19 vaccine and report the rare but serious adverse reactions for early recognition and intervention.
Collapse
Affiliation(s)
- Yuanyuan Yu
- Department of Endocrinology, Air Force Medical Center, Air Force Medical University, Beijing, China
- Department of Endocrinology, Yulin Traditional Chinese Medicine Hospital, Yulin, Shaanxi, China
| | - Guangxin Zhou
- Department of Endocrinology, Air Force Medical Center, Air Force Medical University, Beijing, China
| | - Junjie Du
- Department of Orthopedics, Air Force Medical Center, Air Force Medical University, Beijing, China
| | - Huijuan Zhu
- Department of Endocrinology, Air Force Medical Center, Air Force Medical University, Beijing, China
| | - Haojun Guan
- Department of Endocrinology, Air Force Medical Center, Air Force Medical University, Beijing, China
| | - Yongmin Bi
- Department of Nuclear Medicine, Air Force Medical Center, Air Force Medical University, Beijing, China
| | - Da Zhang
- Department of Endocrinology, Air Force Medical Center, Air Force Medical University, Beijing, China
| |
Collapse
|
43
|
Thankamani K, Shubham D, Kandpal G, Isaac AM, Kavitha MS, Raj VS. Middle East respiratory syndrome coronavirus (MERS-CoV) internalization does not rely on DPP4 cytoplasmic tail signaling. NPJ VIRUSES 2024; 2:67. [PMID: 40295839 PMCID: PMC11721135 DOI: 10.1038/s44298-024-00080-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/05/2024] [Indexed: 04/30/2025]
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) infects respiratory epithelial cells in humans and camels by binding to dipeptidyl peptidase 4 (DPP4) as its entry receptor. DPP4 is a multifunctional type II membrane protein with a long ectodomain and a short six-amino-acid (aa) cytoplasmic tail. MERS-CoV is known to bind to the ectodomain of DPP4 to gain entry into the host cell. However, the role of the cytoplasmic tail in the entry process remains unclear. Here, we show that mutating or deleting individual aa residues or the entire cytoplasmic tail of DPP4 (ΔcytDPP4) does not completely prevent DPP4 from being inserted into the membrane or from allowing the binding of the MERS-CoV spike protein and pseudovirus infection. Although two mutants, ΔcytDPP4, and a single aa deleted DPP4 (ΔK6DPP4) displayed less surface presentation than wtDPP4, the spike protein could still bind and localize on different DPP4 mutants. The reduced surface expression of ΔK6DPP4 might be due to the extended transmembrane domain, which is altered by the hydrophobic tryptophan (W) residue adjacent to the deleted K6. Furthermore, HEK293T cells transiently expressing DPP4 mutants were permeable to MERS-CoV pseudovirus infection. Not only transiently expressing cells but also cells stably expressing the ΔcytDPP4 mutant were susceptible to MERS-CoV pseudoviral infection, indicating that the DPP4 cytoplasmic tail is not required for MERS-CoV entry. Overall, these data suggest that, although MERS-CoV binds to DPP4, other host factors may need to interact with DPP4 or the spike protein to trigger internalization.
Collapse
Affiliation(s)
- Karthika Thankamani
- Virology Scientific Research (VSR) Laboratory, School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER-TVM), Thiruvananthapuram, Kerala, India
| | - Divakar Shubham
- Virology Scientific Research (VSR) Laboratory, School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER-TVM), Thiruvananthapuram, Kerala, India
| | - Gayatri Kandpal
- Virology Scientific Research (VSR) Laboratory, School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER-TVM), Thiruvananthapuram, Kerala, India
| | - Ann Mary Isaac
- Virology Scientific Research (VSR) Laboratory, School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER-TVM), Thiruvananthapuram, Kerala, India
| | - Modenkattil Sethumadhavan Kavitha
- Virology Scientific Research (VSR) Laboratory, School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER-TVM), Thiruvananthapuram, Kerala, India
| | - V Stalin Raj
- Virology Scientific Research (VSR) Laboratory, School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER-TVM), Thiruvananthapuram, Kerala, India.
| |
Collapse
|
44
|
Subbarayan K, Al-Samadi A, Schäfer H, Massa C, Salo T, Biehl K, Vaxevanis CK, Ulagappan K, Wahbi W, Reimers M, Drexler F, Moreira-Soto A, Bachmann M, Seliger B. Altered ACE2 and interferon landscape in the COVID-19 microenvironment correlate with the anti-PD-1 response in solid tumors. Cell Mol Life Sci 2024; 81:473. [PMID: 39625479 PMCID: PMC11615173 DOI: 10.1007/s00018-024-05520-9] [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: 06/21/2024] [Revised: 10/03/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024]
Abstract
Angiotensensin-converting enzyme-2 (ACE2) is a receptor for SARS-CoV-2, allowing the virus to enter cells. Although tumor patients infected by SARS-CoV-2 often have a worse outcome, the expression, function and clinical relevance of ACE2 in tumors has not yet been thoroughly analyzed. In this study, RNA sequencing (RNA-seq) data from tumors, adjacent tissues and whole blood samples of COVID-19 patients from genome databases and from tumor cell lines and endothelial cells infected with different SARS-CoV-2 variants or transfected with an ACE2 expression vector (ACE2high) or mock (ACE2low) were analyzed for the expression of ACE2 and immune response relevant molecules in silico or by qPCR, flow cytometry, Western blot and/or RNA-seq. The differential expression profiles in ACE2high vs. ACE2low cells correlated with available SARS-CoV-2 RNA-seq datasets. ACE2high cells demonstrated upregulated mRNA and/or protein levels of HLA class I, programmed death ligand 1 (PD-L1), components of the antigen processing machinery (APM) and the interferon (IFN) signaling pathway compared to ACE2low cells. Co-cultures of ACE2high cells with peripheral blood mononuclear cells increased immune cell migration and infiltration towards ACE2high cells, apoptosis of ACE2high cells, release of innate immunity-related cytokines and altered NK cell-mediated cytotoxicity. Thus, ACE2 expression was associated in different model systems and upon SARS-CoV-2 infection with an altered host immunogenicity, which might influence the efficacy of immune checkpoint inhibitors. These results provide novel insights into the (patho)physiological role of ACE2 on immune response-relevant mechanisms and suggest an alternative strategy to reduce COVID-19 severity in infected tumor patients targeting the ACE2-induced IFN-PD-L1 axis.
Collapse
Affiliation(s)
- Karthikeyan Subbarayan
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Ahmed Al-Samadi
- Institute of Dentistry, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Joensuu, Finland
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
| | - Helene Schäfer
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Chiara Massa
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
- Institute of Translational Immunology, Brandenburg an der Havel, Germany
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, 90014, Finland
| | - Katharina Biehl
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Christoforos K Vaxevanis
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Kamatchi Ulagappan
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Wafa Wahbi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
| | - Matthias Reimers
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | | | | | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Barbara Seliger
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany.
- Institute of Translational Immunology, Brandenburg an der Havel, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.
- Institute of Translational Medicine, Medical School Theodor Fontane, Hochstr. 29, 14770, Brandenburg an der Havel, Germany.
| |
Collapse
|
45
|
Maiti AK. MDA5 Is a Major Determinant of Developing Symptoms in Critically Ill COVID-19 Patients. Clin Rev Allergy Immunol 2024; 67:58-72. [PMID: 39460899 DOI: 10.1007/s12016-024-09008-z] [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: 10/12/2024] [Indexed: 10/28/2024]
Abstract
Apart from the skin and mucosal immune barrier, the first line of defense of the human immune system includes MDA5 (ifih1 gene) which acts as a cellular sensor protein for certain viruses including SARS-CoV-2. Upon binding with viral RNA, MDA5 activates cell-intrinsic innate immunity, humoral responses, and MAVS (mitochondrial antiviral signaling). MAVS signaling induces type I and III interferon (IFN) expressions that further induce ISGs (interferon stimulatory genes) expressions to initiate human cell-mediated immune responses and attenuate viral replication. SARS-CoV-2 counteracts by producing NSP1, NSP2, NSP3, NSP5, NSP7, NSP12, ORF3A, ORF9, N, and M protein and directs anti-MDA5 antibody production presumably to antagonize IFN signaling. Furthermore, COVID-19 resembles several diseases that carry anti-MDA5 antibodies and the current COVID-19 vaccines induced anti-MDA5 phenotypes in healthy individuals. GWAS (genome-wide association studies) identified several polymorphisms (SNPs) in the ifih1-ifn pathway genes including rs1990760 in ifih1 that are strongly associated with COVID-19, and the associated risk allele is correlated with reduced IFN production. The genetic association of SNPs in ifih1 and ifih1-ifn pathway genes reinforces the molecular findings of the critical roles of MDA5 in sensing SARS-CoV-2 and subsequently the IFN responses to inhibit viral replication and host immune evasion. Thus, MDA5 or its pathway genes could be targeted for therapeutic development of COVID-19.
Collapse
Affiliation(s)
- Amit K Maiti
- Mydnavar, Department of Genetics and Genomics, 28475 Greenfield Rd, Southfield, MI, USA.
| |
Collapse
|
46
|
Megha KB, Reshma S, Amir S, Krishnan MJA, Shimona A, Alka R, Mohanan PV. Comprehensive Risk Assessment of Infection Induced by SARS-CoV-2. Mol Neurobiol 2024; 61:9851-9872. [PMID: 37817031 DOI: 10.1007/s12035-023-03682-4] [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: 06/19/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023]
Abstract
The pandemic COVID-19 (coronavirus disease 2019) is caused by the severe acute respiratory syndrome corona virus 2 (SARS-CoV-2), which devastated the global economy and healthcare system. The infection caused an unforeseen rise in COVID-19 patients and increased the mortality rate globally. This study gives an overall idea about host-pathogen interaction, immune responses to COVID-19, recovery status of infection, targeted organs and complications associated, and comparison of post-infection immunity in convalescent subjects and non-infected individuals. The emergence of the variants and episodes of COVID-19 infections made the situation worsen. The timely introduction of vaccines and precautionary measures helped control the infection's severity. Later, the population that recovered from COVID-19 grew significantly. However, understanding the impact of healthcare issues resulting after infection is paramount for improving an individual's health status. It is now recognised that COVID-19 infection affects multiple organs and exhibits a broad range of clinical manifestations. So, post COVID-19 infection creates a high risk in individuals with already prevailing health complications. The identification of post-COVID-19-related health issues and their appropriate management is of greater importance to improving patient's quality of life. The persistence, sequelae and other medical complications that normally last from weeks to months after the recovery of the initial infection are involved with COVID-19. A multi-disciplinary approach is necessary for the development of preventive measures, techniques for rehabilitation and strategies for clinical management when it comes to long-term care.
Collapse
Affiliation(s)
- K B Megha
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - S Reshma
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - S Amir
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - M J Ajai Krishnan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - A Shimona
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - Rao Alka
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - P V Mohanan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India.
| |
Collapse
|
47
|
Zollner A, Meyer M, Jukic A, Adolph T, Tilg H. The Intestine in Acute and Long COVID: Pathophysiological Insights and Key Lessons. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2024; 97:447-462. [PMID: 39703608 PMCID: PMC11650913 DOI: 10.59249/pmie8461] [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] [Indexed: 12/21/2024]
Abstract
Post-Acute Sequelae of SARS-CoV-2 infection (PASC), commonly known as Long COVID, represents a significant and complex health challenge with a wide range of symptoms affecting multiple organ systems. This review examines the emerging evidence suggesting a critical role of the gut and gut-brain axis in the pathophysiology of Long COVID. It explores how changes in the gut microbiome, disruption of gut barrier integrity, and the persistence of SARS-CoV-2 antigens within the gastrointestinal tract may contribute to the prolonged and varied symptoms seen in Long COVID, including chronic inflammation and neuropsychiatric disturbances. The review also summarizes key insights gained about Long COVID, highlighting its multifactorial nature, which involves immune dysregulation, microvascular damage, and autonomic nervous system dysfunction, with the gut playing a central role in these processes. While progress has been made in understanding these mechanisms, current evidence remains inconclusive. The challenges of establishing causality, standardizing research methodologies, and addressing individual variations in the microbiome are discussed, emphasizing the need for further longitudinal studies and more comprehensive approaches to enhance our understanding of these complex interactions. This review underscores the importance of personalized approaches in developing effective diagnostic and therapeutic strategies for Long COVID, while also acknowledging the significant gaps in our current understanding. Future research should aim to further unravel the complex interplay between the gut and Long COVID, ultimately improving outcomes for those affected by this condition.
Collapse
Affiliation(s)
- Andreas Zollner
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology
& Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Moritz Meyer
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology
& Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Almina Jukic
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology
& Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Timon Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology
& Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology
& Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
48
|
Kim SK, Sung E, Lim K. Recent advances and applications of human lung alveolar organoids. Mol Cells 2024; 47:100140. [PMID: 39490990 PMCID: PMC11629183 DOI: 10.1016/j.mocell.2024.100140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
The human lung alveolus is a well-structured and coordinated pulmonary unit, allowing them to perform diverse functions. While there has been significant progress in understanding the molecular and cellular mechanisms behind human alveolar development and pulmonary diseases, the underlying mechanisms of alveolar differentiation and disease development are still unclear, mainly due to the limited availability of human tissues and a lack of proper in vitro lung model systems mimicking human lung physiology. In this review, we summarize recent advances in creating human lung organoid models that mimic alveolar epithelial cell types. Moreover, we discuss how lung alveolar organoid systems are being applied to recent cutting-edge research on lung development, regeneration, and diseases.
Collapse
Affiliation(s)
- Sun Kyung Kim
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, South Korea
| | - Eunho Sung
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, South Korea
| | - Kyungtae Lim
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, South Korea.
| |
Collapse
|
49
|
Lira Tenório MD, Dos Santos Menezes Siqueira GV, Costa Caldas G, Pacheco de Almeida R, Ribeiro de Jesus A, Martins-Filho PR. Asthma as a risk factor and allergic rhinitis as a protective factor for COVID-19 severity: a case-control study. Eur Arch Otorhinolaryngol 2024; 281:6677-6686. [PMID: 39180537 DOI: 10.1007/s00405-024-08893-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/03/2024] [Indexed: 08/26/2024]
Abstract
PURPOSE The COVID-19 pandemic has resulted in significant global morbidity and mortality. The disease presents a broad clinical spectrum, significantly influenced by underlying comorbidities. While certain conditions are known to exacerbate COVID-19 outcomes, the role of chronic inflammatory airway diseases such as asthma and rhinitis in influencing disease severity remains controversial. This study investigates the association between asthma and allergic rhinitis and the severity of COVID-19 outcomes in a specific geographical region prior to widespread vaccine deployment. METHODS We conducted a case-control study with unvaccinated adult patients who had laboratory-confirmed COVID-19 by polymerase chain reaction (PCR). Cases were defined as severe or critical COVID-19 patients requiring intensive care unit (ICU) admission, and controls were non-severe patients without signs of viral pneumonia or hypoxia. We utilized the International Study of Asthma and Allergies in Childhood (ISAAC) questionnaire to assess the presence of asthma and allergic rhinitis. The association between these chronic inflammatory airway diseases and the severity of COVID-19 was evaluated using multivariate logistic regression analysis. RESULTS A total of 122 patients were analyzed, with 61 in each group. The presence of asthma (9 patients) was associated with an increased likelihood of severe COVID-19 (OR = 13.0; 95% CI 1.27-133.74), while rhinitis (39 patients) was associated with a protective effect against severe outcomes (OR = 0.36; 95% CI 0.13-0.99). No significant association was found between the frequency of asthmatic episodes or the severity of rhinitis and the severity of COVID-19 outcomes. CONCLUSION This study underscores the divergent effects of chronic inflammatory airway diseases on COVID-19 severity, with asthma associated with a higher likelihood of severe outcomes and rhinitis potentially offering protective effects. These findings enhance our understanding of the complex interactions between respiratory allergies and COVID-19, emphasizing the importance of targeted clinical management and public health strategies.
Collapse
Affiliation(s)
- Martha Débora Lira Tenório
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, SE, Brazil
- Investigative Pathology Laboratory, Federal University of Sergipe, Aracaju, SE, Brazil
| | | | | | - Roque Pacheco de Almeida
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, SE, Brazil
- Department of Medicine, Federal University of Sergipe, Aracaju, SE, Brazil
| | - Amélia Ribeiro de Jesus
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, SE, Brazil
- Department of Medicine, Federal University of Sergipe, Aracaju, SE, Brazil
| | - Paulo Ricardo Martins-Filho
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, SE, Brazil.
- Investigative Pathology Laboratory, Federal University of Sergipe, Aracaju, SE, Brazil.
- Hospital Universitário, Laboratório de Patologia Investigativa, Universidade Federal de Sergipe, Rua Cláudio Batista, s/n. Sanatório, Aracaju, CEP: 49060-100, Sergipe, Brasil.
| |
Collapse
|
50
|
Evers P, Uguccioni SM, Ahmed N, Francis ME, Kelvin AA, Pezacki JP. miR-24-3p Is Antiviral Against SARS-CoV-2 by Downregulating Critical Host Entry Factors. Viruses 2024; 16:1844. [PMID: 39772154 PMCID: PMC11680362 DOI: 10.3390/v16121844] [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: 09/23/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Despite all the progress in treating SARS-CoV-2, escape mutants to current therapies remain a constant concern. Promising alternative treatments for current and future coronaviruses are those that limit escape mutants by inhibiting multiple pathogenic targets, analogous to the current strategies for treating HCV and HIV. With increasing popularity and ease of manufacturing of RNA technologies for vaccines and drugs, therapeutic microRNAs represent a promising option. In the present work, miR-24-3p was identified to inhibit SARS-CoV-2 entry, replication, and production; furthermore, this inhibition was retained against common mutations improving SARS-CoV-2 fitness. To determine the mechanism of action, bioinformatic tools were employed, identifying numerous potential effectors promoting infection targeted by miR-24-3p. Of these targets, several key host proteins for priming and facilitating SARS-CoV-2 entry were identified: furin, NRP1, NRP2, and SREBP2. With further experimental analysis, we show that miR-24-3p directly downregulates these viral entry factors to impede infection when producing virions and when infecting the target cell. Furthermore, we compare the findings with coronavirus, HCoV-229E, which relies on different factors strengthening the miR-24-3p mechanism. Taken together, the following work suggests that miR-24-3p could be an avenue to treat current coronaviruses and those likely to emerge.
Collapse
Affiliation(s)
- Parrish Evers
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N6, Canada; (P.E.); (S.M.U.)
| | - Spencer M. Uguccioni
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N6, Canada; (P.E.); (S.M.U.)
| | - Nadine Ahmed
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N6, Canada; (P.E.); (S.M.U.)
| | - Magen E. Francis
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; (M.E.F.); (A.A.K.)
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Alyson A. Kelvin
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; (M.E.F.); (A.A.K.)
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - John P. Pezacki
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N6, Canada; (P.E.); (S.M.U.)
| |
Collapse
|