1
|
Yendewa GA, Olasehinde T, Mulindwa F, Salata RA, Mohareb AM, Jacobson JM. Chronic Hepatitis B and COVID-19 Clinical Outcomes in the United States: A Multisite Retrospective Cohort Study. Open Forum Infect Dis 2025; 12:ofaf013. [PMID: 39896985 PMCID: PMC11786054 DOI: 10.1093/ofid/ofaf013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 01/08/2025] [Indexed: 02/04/2025] Open
Abstract
Background There is conflicting evidence regarding the impact of chronic hepatitis B virus (HBV) on SARS-CoV-2 outcomes. Additionally, the impact of SARS-CoV-2 vaccination and variant periods on outcomes in HBV/SARS-CoV-2 coinfection remain unexplored. Methods We utilized the TriNetX database to compare adults with HBV/SARS-CoV-2 (vs SARS-CoV-2 alone) across 97 US healthcare systems from 2020 to 2023. We assessed the odds of all inpatient hospitalizations, intensive care unit admissions, mechanical ventilation, 30-day, 90-day, and overall mortality. In sensitivity analyses, we excluded HIV, hepatitis C virus, and transplant cases and stratified the HBV/SARS-CoV-2 cohort by cirrhosis status. We applied propensity score matching to address confounding and reported odds ratios (OR) with 95% confidence intervals (CI). Results Of 4 206 774 individuals with SARS-CoV-2, about 0.2% (8293) were HBV/SARS-CoV-2. Individuals with HBV/SARS-CoV-2 (vs SARS-CoV-2 alone) had higher odds of intensive care unit admissions (OR, 1.18; 95% CI, 1.02-1.36), 90-day (OR, 1.22; 95% CI, 1.01-1.41) and overall mortality (OR, 1.18; 95% CI, 1.06-1.33). In sensitivity analyses, those with HBV/SARS-CoV-2 and cirrhosis had a 2.0- to 2.50-fold higher odds of adverse outcomes. Notably, even individuals with HBV/SARS-CoV-2 without cirrhosis had higher odds of mortality. Vaccinated (vs unvaccinated) individuals with HBV/SARS-CoV-2 had 57%, 54%, and 29% reduction in 30-day, 90-day, and overall mortality, respectively. The pre-Delta variant period was associated with higher odds of hospitalization compared to the Omicron but not the Delta period. Conclusions Chronic HBV was associated with worse SARS-CoV-2 outcomes, whereas SARS-CoV-2 vaccination reduced the likelihood of adverse outcomes.
Collapse
Affiliation(s)
- George A Yendewa
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Division of Infectious Diseases and HIV Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Temitope Olasehinde
- Division of Infectious Diseases and HIV Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Frank Mulindwa
- Department of Medicine, United Health Services Wilson Medical Center, Johnson City, New York, USA
| | - Robert A Salata
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Division of Infectious Diseases and HIV Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Amir M Mohareb
- Center for Global Health, Massachusetts General Hospital, Boston, Massachusetts, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey M Jacobson
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Division of Infectious Diseases and HIV Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
2
|
Guironnet-Paquet A, Hamzeh-Cognasse H, Berard F, Cognasse F, Richard JC, Yonis H, Mezidi M, Desebbe O, Delannoy B, Demeret S, Marois C, Saheb S, Le QV, Schoeffler M, Pugliesi PS, Debord S, Bastard P, Cobat A, Casanova JL, Pescarmona R, Viel S, Nicolas JF, Nosbaum A, Vocanson M, Hequet O. Therapeutic plasma exchange accelerates immune cell recovery in severe COVID-19. Front Immunol 2025; 15:1492672. [PMID: 39896810 PMCID: PMC11782122 DOI: 10.3389/fimmu.2024.1492672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/04/2024] [Indexed: 02/04/2025] Open
Abstract
Background Immunological disturbances (anti-type I IFN auto-antibody production, cytokine storm, lymphopenia, T-cell hyperactivation and exhaustion) are responsible for disease exacerbation during severe COVID-19 infections. Methods In this study, we set up a prospective, randomised clinical trial (ClinicalTrials.gov ID: NCT04751643) and performed therapeutic plasma exchange (TPE) in severe COVID-19 patients in order to decrease excess cytokines and auto-antibodies and to assess whether adding TPE to the standard treatment (ST, including corticosteroids plus high-flow rate oxygen) could help restore immune parameters and limit the progression of acute respiratory distress syndrome (ARDS). Results As expected, performing TPE decreased the amount of anti-type I IFN auto-antibodies and improved the elimination or limited the production of certain inflammatory mediators (IL-18, IL-7, CCL2, CCL3, etc.) circulating in the blood of COVID-19 patients, compared to ST controls. Interestingly, while TPE did not influence changes in ARDS parameters throughout the protocol, it proved more effective than ST in reversing lymphopenia, preventing T-cell hyperactivation and reducing T-cell exhaustion, notably in a fraction of TPE patients who had an early favourable respiratory outcome. TPE also restored appropriate numbers of CD4+ and CD8+ T-cell memory populations and increased the number of circulating virus-specific T cells in these patients. Conclusion Our results therefore indicate that the addition of TPE sessions to the standard treatment accelerates immune cell recovery and contributes to the development of appropriate antiviral T-cell responses in some patients with severe COVID-19 disease.
Collapse
Affiliation(s)
- Aurelie Guironnet-Paquet
- Apheresis Unit, Etablissement Français du Sang Auvergne-Rhône-Alpes, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre Bénite, France
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| | - Hind Hamzeh-Cognasse
- University of Jean Monnet, Mines Saint-Étienne, Institut National de la Santé et de la Recherche Médicale (INSERM), U 1059 SAINBIOSE, Saint-Étienne, France
| | - Frederic Berard
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Fabrice Cognasse
- University of Jean Monnet, Mines Saint-Étienne, Institut National de la Santé et de la Recherche Médicale (INSERM), U 1059 SAINBIOSE, Saint-Étienne, France
- Scientific Department, Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France
| | - Jean Christophe Richard
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Hodane Yonis
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Mehdi Mezidi
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Olivier Desebbe
- Department of Anesthesiology and Perioperative Medicine, Sauvegarde Clinic, Ramsay Santé, Lyon, France
| | - Bertrand Delannoy
- Department of Anesthesiology and Perioperative Medicine, Sauvegarde Clinic, Ramsay Santé, Lyon, France
| | - Sophie Demeret
- Neuro-Intensive Care Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Clemence Marois
- Neuro-Intensive Care Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, Institut du Cerveau et de la Moelle (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Departement Médico-Universitaire (DMU) Neurosciences 6, Paris, France
- Groupe de Recherche Clinique en REanimation et Soins Intensifs du Patient en Insuffisance Respiratoire aiguE (GRC-RESPIRE), Sorbonne Université, Paris, France
| | - Samir Saheb
- Hemobiotherapy Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Quoc Viet Le
- Intensive Care Unit, Medipôle Lyon Villeurbanne, Villeurbanne, France
| | - Mathieu Schoeffler
- Department of Anesthesiology and Intensive Care Unit, Centre Hospitalier de Montélimar, Montélimar, France
| | - Paul Simon Pugliesi
- Intensive Care Unit, Centre Hospitalier William Morey, Chalon sur Saône, France
| | - Sophie Debord
- Department of Anesthesiology and Intensive Care Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon (HCL), Lyon, France
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique des Hopitaux de Paris (AP-HP), Paris, France
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
| | - Jean Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique des Hopitaux de Paris (AP-HP), Paris, France
- Howards Hugues Medical Institute, New York, NY, United States
| | - Rémi Pescarmona
- Immun Monitorage Laboratory, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Sébastien Viel
- Plateforme de Biothérapies et de production de Médicaments de Thérapie Innovante (MTI), Hôpital Edouard Herriot, Hospices Civils de Lyon (HCL), Lyon, France
| | - Jean François Nicolas
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Audrey Nosbaum
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Marc Vocanson
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| | - Olivier Hequet
- Apheresis Unit, Etablissement Français du Sang Auvergne-Rhône-Alpes, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre Bénite, France
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| |
Collapse
|
3
|
Paules CI, Nordwall JA, Shaw-Saliba K, Aberg JA, Gardner EM, Goodman AL, Kumarasamy N, Vasudeva S, Vock DM, North CM, Lundgren J, Aggarwal NR. Blood absolute lymphocyte count and trajectory are important in understanding severe COVID-19. BMC Infect Dis 2025; 25:67. [PMID: 39810077 PMCID: PMC11734232 DOI: 10.1186/s12879-024-10428-7] [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: 09/05/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Low blood absolute lymphocyte count (ALC) may predict severe COVID-19 outcomes. Knowledge gaps remain regarding the relationship of ALC trajectory with clinical outcomes and factors associated with lymphopenia. METHODS Our post hoc analysis of the Therapeutics for Inpatients with COVID-19 platform trial utilized proportional hazards models to assess relationships between Day (D) 0 lymphopenia (ALC < 0.9 cells/uL), D0 severe lymphopenia (ALC < 0.5 cells/uL) or lymphopenia trajectory between D0 and D5 with mortality and secondary infections, and with sustained recovery using Fine-Gray models. Logistic regression was used to assess relationships between clinical variables and D0 lymphopenia or lymphopenia trajectory. RESULTS D0 lymphopenia (1426/2579) and severe lymphopenia (636/2579) were associated with increased mortality (aHR 1.48; 1.08, 2.05, p = 0.016 and aHR 1.60; 1.20, 2.14, p = 0.001) and decreased recovery (aRRR 0.90; 0.82, 0.99, p = 0.033 and aRRR 0.78; 0.70, 0.87, p < 0.001 respectively). Trial participants with persistent D5 lymphopenia had increased mortality, and increased secondary infections, and participants with persistent or new lymphopenia had impaired recovery, as compared to participants with no lymphopenia. Persistent and new lymphopenia were associated with older age, male sex; prior immunosuppression, heart failure, aspirin use, and normal body mass index; biomarkers of organ damage (renal and lung), and ineffective immune response (elevated IL-6 and viral nucleocapsid antigen levels). Similar results were observed with severe lymphopenia. CONCLUSIONS Lymphopenia was predictive of severe COVID-19 outcomes, particularly when persistent or new during hospitalization. A better understanding of the underlying risk factors for lymphopenia will help illuminate disease pathogenesis and guide management strategies.
Collapse
Affiliation(s)
- Catharine I Paules
- Division of Infectious Diseases, Penn State Health Milton S. Hershey Medical Center, 500 University Drive, Hershey, PA, 17033, USA.
| | - Jacqueline A Nordwall
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Kathryn Shaw-Saliba
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Judith A Aberg
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Anna L Goodman
- MRC Clinical Trials Unit at University College London and CIDR, King's College London and Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - N Kumarasamy
- VHS Infectious Diseases Medical Centre, CART Clinical Research Site, Voluntary Health Services, Chennai, India
| | - Shikha Vasudeva
- Division of Infectious Diseases, VA Medical Center, Salem, VA, USA
| | - David M Vock
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Crystal M North
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jens Lundgren
- CHIP Center of Excellence for Health, Immunity, and Infections, Department of Infectious Diseases, University of Copenhagen, Righospitalet, Copenhagen, Denmark
| | - Neil R Aggarwal
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
4
|
dos Santos Brito WR, de Brito WB, dos Santos Ferreira F, Santana EGM, da Costa Lopes J, da Silva Graça Amoras E, Lima SS, dos Santos EF, da Costa FP, de Sarges KML, Cantanhede MHD, de Brito MTFM, da Silva ALS, de Meira Leite M, de Nazaré do Socorro de Almeida Viana M, Rodrigues FBB, da Silva R, Viana GMR, do Socorro Souza Chaves T, de Oliveira Lameira Veríssimo A, da Silva Carvalho M, Henriques DF, da Silva CP, Nunes JAL, Costa IB, Brasil-Costa I, Quaresma JAS, Cayres-Vallinoto IMV, Reis LO, Falcão LFM, dos Santos EJM, Vallinoto ACR, Queiroz MAF. Polymorphisms Influence the Expression of the Fas and FasL Genes in COVID-19. Int J Mol Sci 2025; 26:666. [PMID: 39859379 PMCID: PMC11765610 DOI: 10.3390/ijms26020666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
The apoptotic molecule Fas and its ligand FasL are involved in the process of T-lymphocyte death, which may lead to lymphopenia, a characteristic of severe coronavirus disease 2019 (COVID-19). In this study, we investigated the influence of polymorphisms in the FAS and FASL genes, FAS and FASL gene expression, and plasma cytokine levels on COVID-19 severity and long COVID occurrence. A total of 116 individuals with severe COVID-19 and 254 with the non-severe form of the disease were evaluated. In the post-COVID-19 period, samples from 196 individuals with long COVID and 67 from people who did not have long COVID were included. Genotyping and quantification of gene expression were performed via real-time PCR, and cytokine measurement was performed via flow cytometry. The AA genotype for FAS rs1800682 (A/G) and the TT genotype for FASL rs763110 (C/T) were associated with increased FAS and FASL gene expression, respectively (p < 0.005). Higher plasma IFN-γ levels were associated with higher FAS and FASL gene expression (p < 0.05). Among individuals with non-severe COVID-19, carriers of the AA genotype for FAS rs1800682 (A/G) had higher levels of FAS expression, more symptoms, and higher IFN-γ levels (p < 0.05). No association of the evaluated markers with long COVID were observed. The AA genotype of FAS rs1800682 (A/G) and the TT genotype of FASL rs763110 (C/T) influence the levels of FAS and FASL gene expression. Higher gene expression of FAS and FASL may lead to greater inflammation in COVID-19 patients, with higher levels of IFN-γ and T lymphocyte death.
Collapse
Affiliation(s)
- Wandrey Roberto dos Santos Brito
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - William Botelho de Brito
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Fabiane dos Santos Ferreira
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - Emmanuelle Giuliana Mendes Santana
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Jeferson da Costa Lopes
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - Ednelza da Silva Graça Amoras
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Sandra Souza Lima
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Erika Ferreira dos Santos
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Flávia Póvoa da Costa
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Kevin Matheus Lima de Sarges
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Marcos Henrique Damasceno Cantanhede
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Mioni Thieli Figueiredo Magalhães de Brito
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Andréa Luciana Soares da Silva
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Mauro de Meira Leite
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Maria de Nazaré do Socorro de Almeida Viana
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Fabíola Brasil Barbosa Rodrigues
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Rosilene da Silva
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Giselle Maria Rachid Viana
- Laboratory of Basic Research on Malaria, Parasitology Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (G.M.R.V.); (T.d.S.S.C.)
| | - Tânia do Socorro Souza Chaves
- Laboratory of Basic Research on Malaria, Parasitology Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (G.M.R.V.); (T.d.S.S.C.)
- School of Medicine, Institute of Medical Sciences, Federal University of Pará, Belém 66075-110, Brazil
| | | | | | - Daniele Freitas Henriques
- Arbovirology and Hemorrhagic Fevers Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (D.F.H.); (C.P.d.S.)
| | - Carla Pinheiro da Silva
- Arbovirology and Hemorrhagic Fevers Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (D.F.H.); (C.P.d.S.)
| | - Juliana Abreu Lima Nunes
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (J.A.L.N.); (I.B.C.); (I.B.-C.)
| | - Iran Barros Costa
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (J.A.L.N.); (I.B.C.); (I.B.-C.)
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
| | - Igor Brasil-Costa
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (J.A.L.N.); (I.B.C.); (I.B.-C.)
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
| | - Juarez Antônio Simões Quaresma
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
- Center of Biological and Health Sciences, University of the State of Pará, Belém 66087-670, Brazil;
| | - Izaura Maria Vieira Cayres-Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - Leonardo Oliveira Reis
- UroScience, Faculty of Medical Sciences, State University of Campinas, Campinas 13083-590, Brazil;
- ImmunOncology, Pontifical Catholic University of Campinas, Campinas 13060-904, Brazil
| | - Luiz Fábio Magno Falcão
- Center of Biological and Health Sciences, University of the State of Pará, Belém 66087-670, Brazil;
| | - Eduardo José Melo dos Santos
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Antonio Carlos Rosário Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
| | - Maria Alice Freitas Queiroz
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| |
Collapse
|
5
|
Gimenez S, Hamrouni E, André S, Picard M, Soundaramourty C, Lozano C, Vincent T, Tran TA, Kundura L, Estaquier J, Corbeau P. Monocytic reactive oxygen species-induced T-cell apoptosis impairs cellular immune response to SARS-CoV-2 mRNA vaccine. J Allergy Clin Immunol 2025:S0091-6749(25)00011-9. [PMID: 39800264 DOI: 10.1016/j.jaci.2025.01.003] [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: 08/15/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND We have recently shown that during acute severe coronavirus disease 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein induces a cascade of events resulting in T-cell apoptosis. Indeed, by neutralizing the protease activity of its receptor, angiotensin-converting enzyme 2, S protein induces an increase in circulating angiotensin II (AngII), resulting in monocytic release of reactive oxygen species (ROS) and programmed T-cell death. OBJECTIVE Here, we tested whether SARS-CoV-2 mRNA vaccines, known to cause the circulation of the vaccine antigen, S protein receptor binding domain (RBD), might trigger the same cascade. METHODS We used ELISA to quantify the presence of RBD and AngII in peripheral blood of participants and the presence of IFN-γ in the supernatant of PBMCs exposed to S protein. Monocytic ROS production, T-cell apoptosis, and S protein-induced T-lymphocyte proliferation were measured by flow cytometry, and DNA damage was measured by immunofluorescence. RESULTS In most vaccinees, we observed that the presence of circulating RBD peaked on day 14 and was linked to an increase in AngII plasma levels with a peak on day 28. This increase correlated with the ability of monocytes to produce ROS and to induce ROS-mediated DNA damage in neighboring cells, including PBMCs; CD4+ and CD8+ T-lymphocyte apoptosis; and a poor response to protein S in vitro from both CD4+ and CD8+ T cells. CONCLUSIONS We observed the same cascade of events triggered by the vaccinal antigen as by SARS-CoV-2 infection. This cascade may account for the suboptimal efficiency of mRNA SARS-CoV-2 vaccines in preventing the infection, the limited vaccine memory, and certain side effects. In this model, AngII receptor antagonists and/or antioxidants might improve the performance of the SARS-CoV-2 vaccine. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05655351.
Collapse
Affiliation(s)
- Sandrine Gimenez
- Institute of Human Genetics, Molecular Bases of Human Diseases, UMR9002, CNRS and Montpellier University, Montpellier, France
| | - Emna Hamrouni
- Institute of Human Genetics, Molecular Bases of Human Diseases, UMR9002, CNRS and Montpellier University, Montpellier, France
| | - Sonia André
- Université de Paris, INSERM U1124, Mort Cellulaire dans les Interactions Hôte-Pathogène, Paris, France
| | - Morgane Picard
- Université de Paris, INSERM U1124, Mort Cellulaire dans les Interactions Hôte-Pathogène, Paris, France
| | - Calayselvy Soundaramourty
- Université de Paris, INSERM U1124, Mort Cellulaire dans les Interactions Hôte-Pathogène, Paris, France
| | - Claire Lozano
- Immunology Department, University Hospital, Montpellier, France
| | - Thierry Vincent
- Immunology Department, University Hospital, Montpellier, France; Montpellier University, Montpellier, France
| | - Tu-Anh Tran
- Montpellier University, Montpellier, France; Pediatrics Department, University Hospital, Nîmes, France
| | - Lucy Kundura
- Institute of Human Genetics, Molecular Bases of Human Diseases, UMR9002, CNRS and Montpellier University, Montpellier, France
| | - Jérôme Estaquier
- Université de Paris, INSERM U1124, Mort Cellulaire dans les Interactions Hôte-Pathogène, Paris, France; CHU de Québec-Université Laval Research Center, Department of Microbiology and Immunology, Québec City, Québec, Canada.
| | - Pierre Corbeau
- Institute of Human Genetics, Molecular Bases of Human Diseases, UMR9002, CNRS and Montpellier University, Montpellier, France; Montpellier University, Montpellier, France; Immunology Department, University Hospital, Nîmes, France.
| |
Collapse
|
6
|
Missailidis D, Ebrahimie E, Dehcheshmeh MM, Allan C, Sanislav O, Fisher P, Gras S, Annesley SJ. A blood-based mRNA signature distinguishes people with Long COVID from recovered individuals. Front Immunol 2024; 15:1450853. [PMID: 39691709 PMCID: PMC11649547 DOI: 10.3389/fimmu.2024.1450853] [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: 06/18/2024] [Accepted: 11/13/2024] [Indexed: 12/19/2024] Open
Abstract
Introduction Long COVID is a debilitating condition that lasts for more than three months post-infection by SARS-CoV-2. On average, one in ten individuals infected with SARS CoV- 2 develops Long COVID worldwide. A knowledge gap exists in our understanding of the mechanisms, genetic risk factors, and biomarkers that could be associated with Long COVID. Methods In this pilot study we used RNA-Seq to quantify the transcriptomes of peripheral blood mononuclear cells isolated from COVID-recovered individuals, seven with and seven without Long COVID symptoms (age- and sex-matched individuals), on average 6 months after infection. Results Seventy genes were identified as significantly up- or down-regulated in Long COVID samples, and the vast majority were downregulated. The most significantly up- or downregulated genes fell into two main categories, either associated with cell survival or with inflammation. This included genes such as ICOS (FDR p = 0.024) and S1PR1 (FDR p = 0.019) that were both up-regulated, indicating that a pro-inflammatory state is sustained in Long COVID PBMCs compared with COVID recovered PBMCs. Functional enrichment analysis identified that immune-related functions were expectedly predominant among the up- or down-regulated genes. The most frequently downregulated genes in significantly altered functional categories were two leukocyte immunoglobulin like receptors LILRB1 (FDR p = 0.005) and LILRB2 (FDR p = 0.027). PCA analysis demonstrated that LILRB1 and LILRB2 expression discriminated all of the Long COVID samples from COVID recovered samples. Discussion Downregulation of these inhibitory receptors similarly indicates a sustained pro-inflammatory state in Long COVID PBMCs. LILRB1 and LILRB2 should be validated as prospective biomarkers of Long COVID in larger cohorts, over time and against clinically overlapping conditions.
Collapse
Affiliation(s)
- Daniel Missailidis
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, VIC, Australia
| | - Esmaeil Ebrahimie
- Genomics Research Platform, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
- School of Animal and Veterinary Sciences, Faculty of Sciences, Engineering and Technology, University of Adelaide, Adelaide, SA, Australia
| | - Manijeh Mohammadi Dehcheshmeh
- Genomics Research Platform, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Claire Allan
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, VIC, Australia
| | - Oana Sanislav
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, VIC, Australia
| | - Paul Fisher
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, VIC, Australia
| | - Stephanie Gras
- Infection & Immunity Program, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Bundoora, VIC, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Sarah J. Annesley
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
7
|
Wang K, Nie Y, Maguire C, Syphurs C, Sheen H, Karoly M, Lapp L, Gygi JP, Jayavelu ND, Patel RK, Hoch A, Corry D, Kheradmand F, McComsey GA, Fernandez-Sesma A, Simon V, Metcalf JP, Higuita NIA, Messer WB, Davis MM, Nadeau KC, Kraft M, Bime C, Schaenman J, Erle D, Calfee CS, Atkinson MA, Brackenridge SC, Hafler DA, Shaw A, Rahman A, Hough CL, Geng LN, Ozonoff A, Haddad EK, Reed EF, van Bakel H, Kim-Schultz S, Krammer F, Wilson M, Eckalbar W, Bosinger S, Langelier CR, Sekaly RP, Montgomery RR, Maecker HT, Krumholz H, Melamed E, Steen H, Pulendran B, Augustine AD, Cairns CB, Rouphael N, Becker PM, Fourati S, Shannon CP, Smolen KK, Peters B, Kleinstein SH, Levy O, Altman MC, Iwasaki A, Diray-Arce J, Ehrlich LIR, Guan L. Unraveling SARS-CoV-2 Host-Response Heterogeneity through Longitudinal Molecular Subtyping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624784. [PMID: 39651165 PMCID: PMC11623532 DOI: 10.1101/2024.11.22.624784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Hospitalized COVID-19 patients exhibit diverse immune responses during acute infection, which are associated with a wide range of clinical outcomes. However, understanding these immune heterogeneities and their links to various clinical complications, especially long COVID, remains a challenge. In this study, we performed unsupervised subtyping of longitudinal multi-omics immunophenotyping in over 1,000 hospitalized patients, identifying two critical subtypes linked to mortality or mechanical ventilation with prolonged hospital stay and three severe subtypes associated with timely acute recovery. We confirmed that unresolved systemic inflammation and T-cell dysfunctions were hallmarks of increased severity and further distinguished patients with similar acute respiratory severity by their distinct immune profiles, which correlated with differences in demographic and clinical complications. Notably, one critical subtype (SubF) was uniquely characterized by early excessive inflammation, insufficient anticoagulation, and fatty acid dysregulation, alongside higher incidences of hematologic, cardiac, and renal complications, and an elevated risk of long COVID. Among the severe subtypes, significant differences in viral clearance and early antiviral responses were observed, with one subtype (SubC) showing strong early T-cell cytotoxicity but a poor humoral response, slower viral clearance, and greater risks of chronic organ dysfunction and long COVID. These findings provide crucial insights into the complex and context-dependent nature of COVID-19 immune responses, highlighting the importance of personalized therapeutic strategies to improve both acute and long-term outcomes.
Collapse
|
8
|
Chen CC, Lin YA, Liu KT, Huang CY, Shih CM, Lee YT, Pan JL, Lee AW. Navigating SARS-CoV-2-related immunopathology in Crohn's disease: from molecular mechanisms to therapeutic challenges. Virol J 2024; 21:288. [PMID: 39538233 PMCID: PMC11562311 DOI: 10.1186/s12985-024-02529-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) not only posed major health and economic burdens to international societies but also threatens patients with comorbidities and underlying autoimmune disorders, including Crohn's disease (CD) patients. As the vaccinated population is gradually relieved from the stress of the latest omicron variant of SARS-CoV-2 due to competent immune responses, the anxiety of CD patients, especially those on immunosuppressive treatment, has not subsided. Whether the use of immunosuppressants for remission of CD outweighs the potential risk of severe coronavirus disease 2019 (COVID-19) has long been discussed. Thus, for the best benefit of CD patients, our primary goal in this study was to navigate the clinical management of CD during the COVID pandemic. Herein, we summarized COVID-19 outcomes of CD patients treated with immunosuppressive agents from multiple cohort studies and also investigated possible mechanisms of how SARS-CoV-2 impacts the host immunity with special consideration of CD patients. We first looked into the SARS-CoV-2-related immunopathology, including lymphocytopenia, T-cell exhaustion, cytokine storms, and their possible molecular interactions, and then focused on mechanistic actions of gastrointestinal systems, including interruption of tryptophan absorption, development of dysbiosis, and consequent local and systemic inflammation. Given challenges in managing CD, we summarized up-to-date clinical and molecular evidence to help physicians adjust therapeutic strategies to achieve the best clinical outcomes for CD patients.
Collapse
Affiliation(s)
- Chang-Cyuan Chen
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-An Lin
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kuan-Ting Liu
- Department of General Medicine, Chang Gung Memorial Hospital, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chun-Yao Huang
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chun-Ming Shih
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yuan-Ti Lee
- School of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
- Division of Infectious Diseases, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan
| | - Jun-Liang Pan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, 11031, Taiwan.
| | - Ai-Wei Lee
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
9
|
Ruiz Seco MP, Paño Pardo JR, Schoergenhofer C, Dings C, Lehr T, Herth F, Krendyukov A, Straub C, Kappler M, Jilma B, Fricke H, Pardo J, de Miguel D, Thiemann M, Bergmann M, Walczak H, Hoeger T. Efficacy and safety of asunercept, a CD95L-selective inhibitor, in hospitalised patients with moderate-to-severe COVID-19: ASUNCTIS, a multicentre, randomised, open-label, controlled, phase 2 trial. EClinicalMedicine 2024; 77:102879. [PMID: 39513186 PMCID: PMC11541427 DOI: 10.1016/j.eclinm.2024.102879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 11/15/2024] Open
Abstract
Background The phase 2 ASUNCTIS study assessed the efficacy and safety of asunercept, a fully human CD95 (Fas) ligand-binding protein, in hospitalised patients with moderate-to-severe coronavirus disease (COVID-19) to assess the clinical benefit of CD95 ligand inhibition in this viral disease. Methods In this open-label, multicentre, randomised, controlled, phase 2 trial, patients with COVID-19-induced pneumonia and respiratory deterioration were randomly assigned (1:1:1:1) in 12 Russian and Spanish hospitals using an interactive web-response system to receive standard of care (SOC) or SOC plus weekly asunercept 25 mg, 100 mg, or 400 mg, administered intravenously for up to 4 weeks, or until hospital discharge or death. The randomisation was stratified according to the respiratory support methods at the time of enrolment, corresponding to categories 4-6 of a clinical severity assessment scale comprising 9 levels that was recommended by the World Health Organization (WHO) at the time of the study. The main inclusion criterion was laboratory confirmed infection with SARS-CoV-2 OR typical radiological signs of SARS-CoV-2 infection. The primary endpoint was time from randomisation to clinical improvement on two consecutive days of at least one category on a WHO clinical severity assessment scale in the modified intent-to-treat population. All patients were subjected to regular safety analyses. This trial is registered with EudraCT (2020-001887-27) and ClinicalTrials.gov (NCT04535674). Findings Between October 9, 2020, and September 24, 2021, 438 patients were randomly assigned to SOC (n = 110) or SOC plus asunercept 25 mg (n = 109), 100 mg (n = 109), or 400 mg (n = 110). The primary endpoint, time to sustained clinical improvement of one WHO category on two consecutive days from randomization, was in median [95% confidence interval]: 9 [6-12], 8 [7-12], 8 [7-11] and 13 [9-20] days for the 400 mg, 100 mg, 25 mg asunercept and SOC groups, respectively. The standard deviations for the 400 mg, 100 mg, 25 mg asunercept and SOC groups were 5.3, 4.9, 4.7 and 5 days, respectively. The observed differences between groups failed to reach statistical significance (one-sided p-value = 0.041). In total, 290 adverse events (AE) were registered in 145 patients who received at least one dose of the study treatment: 77 AEs in 37 (33.6%) patients in the SOC group, 80 AEs in 38 (34.9%) patients in the 25 mg group, 61 AEs in 35 (32.7%) patients in the 100 mg group and 72 AEs in 35 (32.1%) patients in the 400 mg group. There was no treatment-related death reported. In summary, asunercept was well tolerated at all doses tested and no specific safety signals were detected. Interpretation The primary endpoint of time to sustained clinical improvement for distinct asunercept arms compared to SOC failed to meet statistical significance. The compound was safe and well tolerated. Funding Apogenix GmbH, Heidelberg, Germany.
Collapse
Affiliation(s)
- Maria Pilar Ruiz Seco
- Department of Internal Medicine, University Hospital Infanta Sofia, Paseo de Europa, 34, 28703, San Sebastián de los Reyes, Madrid, Spain
| | - José Ramón Paño Pardo
- Department of Infectious Diseases, Clinical University Hospital Lozano Blesa/Aragon Health Research Institute (IISA), Avda. San Juan Bosco, 15, 50009 Zaragoza, Spain
- CIBER de Enfermedades Infecciosas, Avda. de Monforte de Lemos, 5, 28029, IS Carlos III, Madrid, Spain
| | - Christian Schoergenhofer
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Christiane Dings
- Department of Clinical Pharmacy, Saarland University, 66123 Saarbrücken, Germany
- Saarmetrics GmbH, Starterzentrum 1, Universität des Saarlandes, 66123 Saarbrücken, Germany
| | - Thorsten Lehr
- Department of Clinical Pharmacy, Saarland University, 66123 Saarbrücken, Germany
- Saarmetrics GmbH, Starterzentrum 1, Universität des Saarlandes, 66123 Saarbrücken, Germany
| | - Felix Herth
- Thoraxklinik, Roentgenstr. 1, 69126 Heidelberg, Germany
| | | | - Carola Straub
- Apogenix GmbH, Im Neuenheimer Feld 584, 69120 Heidelberg, Germany
| | | | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Harald Fricke
- Apogenix GmbH, Im Neuenheimer Feld 584, 69120 Heidelberg, Germany
| | - Julian Pardo
- CIBER de Enfermedades Infecciosas, Avda. de Monforte de Lemos, 5, 28029, IS Carlos III, Madrid, Spain
- Department of Microbiology, Radiology, Paediatric and Public Health, University of Zaragoza/Aragon Health Research Institute (IISA), Domingo Miral s/n, 50009 Zaragoza, Spain
| | - Diego de Miguel
- Department of Microbiology, Radiology, Paediatric and Public Health, University of Zaragoza/Aragon Health Research Institute (IISA), Domingo Miral s/n, 50009 Zaragoza, Spain
| | - Meinolf Thiemann
- Apogenix GmbH, Im Neuenheimer Feld 584, 69120 Heidelberg, Germany
| | - Michael Bergmann
- Division of Visceral Surgery and Comprehensive Cancer Center, Department of General Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Henning Walczak
- Institute of Biochemistry I, Centre for Biochemistry, and CECAD Research Centre, University of Cologne, Joseph-Stelzmann Str. 26, 50931 Cologne, Germany
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, 72 Huntley St, WC1E 6BT London, UK
| | - Thomas Hoeger
- Apogenix GmbH, Im Neuenheimer Feld 584, 69120 Heidelberg, Germany
| |
Collapse
|
10
|
Yang T, Lian H, Liao J, Zeng Y, Li J, Lin C, Lin M. Epidemiological characteristics and meteorological factors of acute respiratory infections (ARIs) in hospitalized children in eastern Guangdong, China. Sci Rep 2024; 14:25518. [PMID: 39462026 PMCID: PMC11513138 DOI: 10.1038/s41598-024-77005-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Acute respiratory infections (ARIs) are the most common issue in pediatric clinical practice. They pose a significant public threat, with high morbidity and mortality rates worldwide. Aiming at understanding the epidemiological characteristics of respiratory pathogens and their risk factors among children in eastern Guangdong, China. Samples obtained from 15,993 children hospitalized with ARIs in eastern Guangdong Province were tested for 14 pathogens via multiplex polymerase chain reaction (PCR) from May 2019 to July 2023. The number of hospitalizations for ARIs was correlated with pathogens, age, meteorological parameters, and the pandemic of COVID-19. The data were analyzed by different statistical methods. Among all the samples, the positive rate with ARIs accounted for 68.94% (11,026/15,993) in hospitalized patients. Cytomegalovirus (CMV) (24.49%), Streptococcus pneumoniae (SP) (20.54%), and Respiratory Syncytial Virus (RSV) (14.16%) were the top three pathogens with the greatest infection rates. Among hospitalized patients, there were more single infections in pediatric patients (40.91%, P < 0.001). Compared with bacterial infection and mixed infection, the detection rate of virus infection was higher in pediatric (36.04%, P < 0.001). Age-related increases in Mycoplasma pneumoniae (MP) infection (r = 0.729, P < 0.001) and decreases in RSV infection were observed (r = 0.88, P < 0.001). The virus infection peaked at six months, and the bacterial infection and mixed infection peaked at 1-3 years. Viral pathogens are on the rise in the post-pandemic era. The prevalence of SP infection was more influenced by the Air Quality Index (AQI), RSV infections were more clearly influenced by temperature, and Influenza A virus (IAV) infections were more strongly correlated with both the AQI and relative humidity (P < 0.001). This study highlights the need of keeping an eye on monitoring meteorological factors in assessing hospitalizations for pediatric ARIs in eastern Guangdong, China, especially RSV- and SP-associated hospitalizations.
Collapse
Affiliation(s)
- Tiandan Yang
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Haobin Lian
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Jiayu Liao
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Yongmei Zeng
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Jiamin Li
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Chuangxing Lin
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515000, Guangdong, China.
| | - Min Lin
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521000, Guangdong, China.
| |
Collapse
|
11
|
Murdocca M, Andrade Santos-Filho O, De Masi C, Dos Santos Rodrigues E, Campos de Souza CV, De Santis R, Amatore D, Latini A, Schipani R, di Rienzo Businco L, Brandimarte B, Grilli G, Huang TL, Mayence AS, Lista F, Duranti A, Sangiuolo F, Vanden Eynde JJ, Novelli G. Characterization of the symmetrical benzimidazole twin drug TL1228: the role as viral entry inhibitor for fighting COVID-19. Biol Direct 2024; 19:93. [PMID: 39415197 PMCID: PMC11481581 DOI: 10.1186/s13062-024-00523-9] [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: 08/14/2024] [Accepted: 08/28/2024] [Indexed: 10/18/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is reliably one of the largest pandemics the world has suffered in recent years. In the search for non-biological antivirals, special emphasis was placed on drug repurposing to accelerate the clinical implementation of effective drugs.The life cycle of the virus has been extensively investigated and many human targets have been identified, such as the molecular chaperone GRP78, representing a host auxiliary factor for SARS-CoV-2 entry. Here we report the inhibitor capacity of TL1228, a small molecule discovered through an in silico screening approach, which could interfere with the interaction of SARS-CoV-2 and its target cells, blocking the recognition of the GRP78 cellular receptor by the viral Spike protein. TL1228 showed in vitro the ability to reduce significantly both pseudoviral and authentic viral activity even through the reduction of GRP78/ACE2 transcript levels. Importantly, TL1228 acts in modulating expression levels of innate immunity and as inflammation markers.
Collapse
Affiliation(s)
- Michela Murdocca
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Osvaldo Andrade Santos-Filho
- Center of Health Sciences Laboratory of Molecular Modelling & Computational Strutural Biology Cidade Universitária, Federal University of Rio de Janeiro IPPN, Av. Carlos Chagas Filho373, Bloco H, Rio de Janeiro, 21941-599, RJ, Brazil
| | - Claudia De Masi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Edivaldo Dos Santos Rodrigues
- Center of Health Sciences Laboratory of Molecular Modelling & Computational Strutural Biology Cidade Universitária, Federal University of Rio de Janeiro IPPN, Av. Carlos Chagas Filho373, Bloco H, Rio de Janeiro, 21941-599, RJ, Brazil
| | - Claudia Valeria Campos de Souza
- Center of Health Sciences Laboratory of Molecular Modelling & Computational Strutural Biology Cidade Universitária, Federal University of Rio de Janeiro IPPN, Av. Carlos Chagas Filho373, Bloco H, Rio de Janeiro, 21941-599, RJ, Brazil
| | - Riccardo De Santis
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy
- Defence Institute for Biomedical Sciences, Rome, 00184, Italy
| | | | - Andrea Latini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Rossella Schipani
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Lino di Rienzo Businco
- Otorhinolaryngology Department, Institute of Sport Medicine and Science CONI, Rome, Italy
| | - Bruno Brandimarte
- Electronic Measurements Physics Department, Sapienza University, Rome, Italy
| | - Giorgia Grilli
- Defence Institute for Biomedical Sciences, Rome, 00184, Italy
| | - Tien L Huang
- Formerly Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA, 70125, USA
| | - Annie S Mayence
- Formerly Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA, 70125, USA
| | - Florigio Lista
- Defence Institute for Biomedical Sciences, Rome, 00184, Italy
| | - Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, 61029, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| | - Jean Jacques Vanden Eynde
- Formerly Department of Organic Chemistry (FS), University of Mons-UMONS, 1 place du Parc, Mons, 7000, Belgium
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
12
|
Bispo ECI, Argañaraz ER, Neves FDAR, de Carvalho JL, Saldanha-Araujo F. Immunomodulatory effect of IFN-γ licensed adipose-mesenchymal stromal cells in an in vitro model of inflammation generated by SARS-CoV-2 antigens. Sci Rep 2024; 14:24235. [PMID: 39415027 PMCID: PMC11484699 DOI: 10.1038/s41598-024-75776-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024] Open
Abstract
In recent years, clinical studies have shown positive results of the application of Mesenchymal Stromal Cells (MSCs) in severe cases of COVID-19. However, the mechanisms of immunomodulation of IFN-γ licensed MSCs in SARS-CoV-2 infection are only partially understood. In this study, we first tested the effect of IFN-γ licensing in the MSC immunomodulatory profile. Then, we established an in vitro model of inflammation by exposing Calu-3 lung cells to SARS-CoV-2 nucleocapsid and spike (NS) antigens, and determined the toxicity of SARS-CoV-2 NS antigen and/or IFN-γ stimulation to Calu-3. The conditioned medium (iCM) generated by Calu-3 cells exposed to IFN-γ and SARS-CoV-2 NS antigens was used to stimulate T-cells, which were then co-cultured with IFN-γ-licensed MSCs. The exposure to IFN-γ and SARS-CoV-2 NS antigens compromised the viability of Calu-3 cells and induced the expression of the inflammatory mediators ICAM-1, CXCL-10, and IFN-β by these cells. Importantly, despite initially stimulating T-cell activation, IFN-γ-licensed MSCs dramatically reduced IL-6 and IL-10 levels secreted by T-cells exposed to NS antigens and iCM. Moreover, IFN-γ-licensed MSCs were able to significantly inhibit T-cell apoptosis induced by SARS-CoV-2 NS antigens. Taken together, our data show that, in addition to reducing the level of critical cytokines in COVID-19, IFN-γ-licensed MSCs protect T-cells from SARS-CoV-2 antigen-induced apoptosis. Such observations suggest that MSCs may contribute to COVID-19 management by preventing the lymphopenia and immunodeficiency observed in critical cases of the disease.
Collapse
Affiliation(s)
- Elizabete Cristina Iseke Bispo
- Laboratory of Hematology and Stem Cells (LHCT), Faculty of Health Sciences, University of Brasília, Brasília, 70910-900, Brazil
| | - Enrique Roberto Argañaraz
- Laboratory of Molecular NeuroVirology, Faculty of Health Sciences, University of Brasília, Brasília, 70910-900, Brazil
| | | | - Juliana Lott de Carvalho
- Interdisciplinary Laboratory of Bioscience, Faculty of Medicine, University of Brasília, Brasília, 70910-900, Brazil
| | - Felipe Saldanha-Araujo
- Laboratory of Hematology and Stem Cells (LHCT), Faculty of Health Sciences, University of Brasília, Brasília, 70910-900, Brazil.
| |
Collapse
|
13
|
Peña-Bates C, Lascurain R, Ortiz-Navarrete V, Chavez-Galan L. The BCG vaccine and SARS-CoV-2: Could there be a beneficial relationship? Heliyon 2024; 10:e38085. [PMID: 39347386 PMCID: PMC11437859 DOI: 10.1016/j.heliyon.2024.e38085] [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: 03/28/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
The COVID-19 disease continues to cause complications and deaths worldwide. Identifying effective immune protection strategies remains crucial to address this ongoing challenge. The Bacillus Calmette-Guérin (BCG) vaccine, developed initially to prevent pulmonary tuberculosis, has gained relevance due to its ability to induce cross-protection against other pathogens of the airways. This review summarizes research on the immunological protection provided by BCG, along with its primary clinical and therapeutic uses. It also explores the immunological features of COVID-19, the mechanisms implicated in host cell death, and its association with chronic pulmonary illnesses such as tuberculosis, which has led to complications in diagnosis and management. While vaccines against COVID-19 have been administered globally, uncertainty still exists about its effectiveness. Additionally, it is uncertain whether the utilization of BCG can regulate the immune response to pathogens such as SARS-CoV-2.
Collapse
Affiliation(s)
- Carlos Peña-Bates
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Ricardo Lascurain
- Unidad de Enlace Científico, Faculty of Medicine, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, 14610, Mexico
| | - Vianney Ortiz-Navarrete
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Leslie Chavez-Galan
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| |
Collapse
|
14
|
Chen WC, Hu SY, Cheng CM, Shen CF, Chuang HY, Ker CR, Sun DJ, Shen CJ. Evaluating TRAIL and IP-10 alterations in vaccinated pregnant women after COVID-19 diagnosis and their correlation with neutralizing antibodies. Front Immunol 2024; 15:1415561. [PMID: 39290698 PMCID: PMC11405216 DOI: 10.3389/fimmu.2024.1415561] [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: 04/11/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024] Open
Abstract
Background This study evaluates tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and interferon-γ-induced protein-10 (IP-10) in pregnant women with COVID-19 and their newborns, exploring the effects of antiviral treatments and vaccine-induced neutralizing antibody (Nab) inhibition on these key viral infection biomarkers. Methods We studied 61 pregnant women with past COVID-19 and either three (n=56) or four (n=5) doses of vaccination, and 46 without COVID-19 but vaccinated. We analyzed them and their newborns' blood for TRAIL, IP-10, and Nab levels using enzyme-linked immunosorbent assays (ELISA), correlating these with other clinical factors. Results Our study found lower TRAIL but higher IP-10 levels in maternal blood than neonatal cord blood, irrespective of past COVID-19 diagnosis. Cases diagnosed with COVID-19 < 4 weeks previously had higher maternal blood TRAIL levels (16.49 vs. 40.81 pg/mL, p=0.0064) and IP-10 (154.68 vs. 225.81 pg/mL, p=0.0170) than those never diagnosed. Antiviral medication lowered TRAIL and IP-10 in maternal blood without affecting Nab inhibition (TRAIL: 19.24 vs. 54.53 pg/mL, p=0.028; IP-10: 158.36 vs. 255.47 pg/mL, p=0.0089). TRAIL and IP-10 levels were similar with three or four vaccine doses, but four doses increased Nab inhibition (p=0.0363). Previously COVID-19 exposed pregnant women had higher Nab inhibition (p < 0.0001). No obvious correlation was found among TRAIL, IP-10, and Nab inhibition level. Conclusions Our study suggests that lower maternal TRAIL and higher IP-10 levels compared to neonatal cord blood coupled with a rise in both markers following COVID-19 diagnosis that could be reduced by antivirals indicates a correlation to infection severity. Higher vaccine doses enhance Nab inhibition, irrespective of antiviral medication use and independent of TRAIL or IP-10 levels, highlighting the significance and safety of adequate vaccination and antiviral use post-diagnosis in pregnant women.
Collapse
Affiliation(s)
- Wei-Chun Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Obstetrics and Gynecology, New Taipei City Municipal Tucheng Hospital, New Taipei City, Taiwan
- International Intercollegiate Ph.D. Program, National Tsing Hua University, Hsinchu, Taiwan
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shu-Yu Hu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Fen Shen
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Yu Chuang
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin-Ru Ker
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Der-Ji Sun
- Department of Obstetrics and Gynecology, Pojen Hospital, Kaohsiung, Taiwan
| | - Ching-Ju Shen
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
15
|
Yu J, Zhang Y, Ye Z, Tang K, Ma Y, Fu L, Cui T, Kang H, Yuan Y, Pan W. A Multi-Machine Learning Consensus Model Based on Clinical Features Reveals That Interleukin-10 Derived from Monocytes Leads to a Poor Prognosis in Patients with Coronavirus Disease-2019. J Inflamm Res 2024; 17:5923-5942. [PMID: 39247837 PMCID: PMC11378990 DOI: 10.2147/jir.s472099] [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: 06/03/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024] Open
Abstract
Background Despite ongoing interventions, SARS-CoV-2 continues to cause significant global morbidity and mortality. Early diagnosis and intervention are crucial for effective clinical management. However, prognostic features based on transcriptional data have shown limited effectiveness, highlighting the need for more precise biomarkers to improve COVID-19 treatment outcomes. Methods We retrospectively analyzed 149 clinical features from 189 COVID-19 patients, identifying prognostic features via univariate Cox regression. The cohort was split into training and validation sets, and 77 prognostic models were developed using seven machine learning algorithms. Among these, the least absolute shrinkage and selection operator (Lasso) method was employed to refine the selection of prognostic variables by ten-fold cross-validation strategy, which were then integrated with random survival forests (RSF) to build a robust COVID-19-related prognostic model (CRM). Model accuracy was evaluated across training, validation, and entire cohorts. The diagnostic relevance of interleukin-10 (IL-10) was confirmed in bulk transcriptional data and validated at the single-cell level, where we also examined changes in cellular communication between mononuclear cells with differing IL-10 expression and other immune cells. Results Univariate Cox regression identified 43 prognostic features. Among the 77 machine learning models, the combination of Lasso and RSF produced the most robust CRM. This model consistently performed well across training, validation, and entire cohorts. IL-10 emerged as a key prognostic feature within the CRM, validated by single-cell transcriptional data. Transcriptome analysis confirmed the stable diagnostic value of IL-10, with mononuclear cells identified as the primary IL-10 source. Moreover, differential IL-10 expression in these cells was linked to altered cellular communication in the COVID-19 immune microenvironment. Conclusion The CRM provides accurate prognostic predictions for COVID-19 patients. Additionally, the study underscores the importance of early IL-10 level testing upon hospital admission, which could inform therapeutic strategies.
Collapse
Affiliation(s)
- Jing Yu
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yike Zhang
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Zhixiong Ye
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Kun Tang
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yiming Ma
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Linlin Fu
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Tongtong Cui
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Hening Kang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yadong Yuan
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Wensen Pan
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| |
Collapse
|
16
|
Zhao K, So HC, Lin Z. scParser: sparse representation learning for scalable single-cell RNA sequencing data analysis. Genome Biol 2024; 25:223. [PMID: 39152499 PMCID: PMC11328435 DOI: 10.1186/s13059-024-03345-0] [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: 07/31/2023] [Accepted: 07/23/2024] [Indexed: 08/19/2024] Open
Abstract
The rapid rise in the availability and scale of scRNA-seq data needs scalable methods for integrative analysis. Though many methods for data integration have been developed, few focus on understanding the heterogeneous effects of biological conditions across different cell populations in integrative analysis. Our proposed scalable approach, scParser, models the heterogeneous effects from biological conditions, which unveils the key mechanisms by which gene expression contributes to phenotypes. Notably, the extended scParser pinpoints biological processes in cell subpopulations that contribute to disease pathogenesis. scParser achieves favorable performance in cell clustering compared to state-of-the-art methods and has a broad and diverse applicability.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Statistics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Hon-Cheong So
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology and The Chinese University of Hong Kong, Hong Kong SAR, China.
- Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- Margaret K.L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- Brain and Mind Institute, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- Hong Kong Branch of the Chinese Academy of Sciences Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Zhixiang Lin
- Department of Statistics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
17
|
Peng D, Wang L, Fang Y, Lu L, Li Z, Jiang S, Chen J, Aschner M, Li S, Jiang Y. Lead exposure induces neurodysfunction through caspase-1-mediated neuronal pyroptosis. ENVIRONMENTAL RESEARCH 2024; 255:119210. [PMID: 38795947 DOI: 10.1016/j.envres.2024.119210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024]
Abstract
Chronic lead (Pb) exposure causes neurodysfunction and contributes to the development of neurodegenerative disease. However, the mechanism of Pb-induced neurological dysfunction have yet to be fully elucidated. This study determined the role pyroptosis plays in Pb-induced neurodysfunction in neurons. We used both in vitro and in vivo approaches to explore whether Pb exposure induces caspase-1-mediated pyroptosis in neurons and its relationship to Pb-induced neurological disorders. Our findings showed that caspase-1-mediated pyroptosis in Pb-exposed neurons activated glycogen synthase kinase 3 protease activity by disrupting Ca2+/calmodulin-dependent protein kinase II/cAMP-response element binding protein pathway, leading to neurological disorders. Moreover, the caspase-1 inhibition VX-765 or the non-steroidal anti-inflammatory drug sodium para-aminosalicylic acid (PAS-Na) attenuated the Pb-induced neurological disorders by alleviating caspase-1 mediated neuronal pyroptosis. Our novel studies suggest that caspase-1-mediated pyroptosis in neurons represents a potential mechanism for Pb-induced neurodysfunction, identifying a putative target for attenuating the neurodegenerative effects induced by this metal.
Collapse
Affiliation(s)
- Dongjie Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Leilei Wang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Yuanyuan Fang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Lili Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Zhaocong Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Siyang Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Jing Chen
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Yueming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| |
Collapse
|
18
|
Chidambaram V, Kumar A, Sadaf MI, Lu E, Al’Aref SJ, Tarun T, Galiatsatos P, Gulati M, Blumenthal RS, Leucker TM, Karakousis PC, Mehta JL. COVID-19 in the Initiation and Progression of Atherosclerosis: Pathophysiology During and Beyond the Acute Phase. JACC. ADVANCES 2024; 3:101107. [PMID: 39113913 PMCID: PMC11304887 DOI: 10.1016/j.jacadv.2024.101107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 04/24/2024] [Accepted: 06/01/2024] [Indexed: 08/10/2024]
Abstract
The incidence of atherosclerotic cardiovascular disease is increasing globally, especially in low- and middle-income countries, despite significant efforts to reduce traditional risk factors. Premature subclinical atherosclerosis has been documented in association with several viral infections. The magnitude of the recent COVID-19 pandemic has highlighted the need to understand the association between SARS-CoV-2 and atherosclerosis. This review examines various pathophysiological mechanisms, including endothelial dysfunction, platelet activation, and inflammatory and immune hyperactivation triggered by SARS-CoV-2 infection, with specific attention on their roles in initiating and promoting the progression of atherosclerotic lesions. Additionally, it addresses the various pathogenic mechanisms by which COVID-19 in the post-acute phase may contribute to the development of vascular disease. Understanding the overlap of these syndromes may enable novel therapeutic strategies. We further explore the need for guidelines for closer follow-up for the often-overlooked evidence of atherosclerotic cardiovascular disease among patients with recent COVID-19, particularly those with cardiometabolic risk factors.
Collapse
Affiliation(s)
- Vignesh Chidambaram
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Amudha Kumar
- Division of Cardiology, Department of Medicine, Loyola University Medical Center, Maywood, Illinois, USA
| | - Murrium I. Sadaf
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Emily Lu
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Subhi J. Al’Aref
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Tushar Tarun
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Panagis Galiatsatos
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Martha Gulati
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Roger S. Blumenthal
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thorsten M. Leucker
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Petros C. Karakousis
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jawahar L. Mehta
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Division of Cardiovascular Medicine, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
19
|
Zheng HY, Song TZ, Zheng YT. Immunobiology of COVID-19: Mechanistic and therapeutic insights from animal models. Zool Res 2024; 45:747-766. [PMID: 38894519 PMCID: PMC11298684 DOI: 10.24272/j.issn.2095-8137.2024.062] [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: 02/23/2024] [Accepted: 04/22/2024] [Indexed: 06/21/2024] Open
Abstract
The distribution of the immune system throughout the body complicates in vitro assessments of coronavirus disease 2019 (COVID-19) immunobiology, often resulting in a lack of reproducibility when extrapolated to the whole organism. Consequently, developing animal models is imperative for a comprehensive understanding of the pathology and immunology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This review summarizes current progress related to COVID-19 animal models, including non-human primates (NHPs), mice, and hamsters, with a focus on their roles in exploring the mechanisms of immunopathology, immune protection, and long-term effects of SARS-CoV-2 infection, as well as their application in immunoprevention and immunotherapy of SARS-CoV-2 infection. Differences among these animal models and their specific applications are also highlighted, as no single model can fully encapsulate all aspects of COVID-19. To effectively address the challenges posed by COVID-19, it is essential to select appropriate animal models that can accurately replicate both fatal and non-fatal infections with varying courses and severities. Optimizing animal model libraries and associated research tools is key to resolving the global COVID-19 pandemic, serving as a robust resource for future emerging infectious diseases.
Collapse
Affiliation(s)
- Hong-Yi Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Tian-Zhang Song
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Yong-Tang Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China. E-mail:
| |
Collapse
|
20
|
Agamah FE, Ederveen THA, Skelton M, Martin DP, Chimusa ER, ’t Hoen PAC. Network-based integrative multi-omics approach reveals biosignatures specific to COVID-19 disease phases. Front Mol Biosci 2024; 11:1393240. [PMID: 39040605 PMCID: PMC11260748 DOI: 10.3389/fmolb.2024.1393240] [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: 02/28/2024] [Accepted: 05/22/2024] [Indexed: 07/24/2024] Open
Abstract
Background COVID-19 disease is characterized by a spectrum of disease phases (mild, moderate, and severe). Each disease phase is marked by changes in omics profiles with corresponding changes in the expression of features (biosignatures). However, integrative analysis of multiple omics data from different experiments across studies to investigate biosignatures at various disease phases is limited. Exploring an integrative multi-omics profile analysis through a network approach could be used to determine biosignatures associated with specific disease phases and enable the examination of the relationships between the biosignatures. Aim To identify and characterize biosignatures underlying various COVID-19 disease phases in an integrative multi-omics data analysis. Method We leveraged a multi-omics network-based approach to integrate transcriptomics, metabolomics, proteomics, and lipidomics data. The World Health Organization Ordinal Scale WHO Ordinal Scale was used as a disease severity reference to harmonize COVID-19 patient metadata across two studies with independent data. A unified COVID-19 knowledge graph was constructed by assembling a disease-specific interactome from the literature and databases. Disease-state specific omics-graphs were constructed by integrating multi-omics data with the unified COVID-19 knowledge graph. We expanded on the network layers of multiXrank, a random walk with restart on multilayer network algorithm, to explore disease state omics-specific graphs and perform enrichment analysis. Results Network analysis revealed the biosignatures involved in inducing chemokines and inflammatory responses as hubs in the severe and moderate disease phases. We observed distinct biosignatures between severe and moderate disease phases as compared to mild-moderate and mild-severe disease phases. Mild COVID-19 cases were characterized by a unique biosignature comprising C-C Motif Chemokine Ligand 4 (CCL4), and Interferon Regulatory Factor 1 (IRF1). Hepatocyte Growth Factor (HGF), Matrix Metallopeptidase 12 (MMP12), Interleukin 10 (IL10), Nuclear Factor Kappa B Subunit 1 (NFKB1), and suberoylcarnitine form hubs in the omics network that characterizes the moderate disease state. The severe cases were marked by biosignatures such as Signal Transducer and Activator of Transcription 1 (STAT1), Superoxide Dismutase 2 (SOD2), HGF, taurine, lysophosphatidylcholine, diacylglycerol, triglycerides, and sphingomyelin that characterize the disease state. Conclusion This study identified both biosignatures of different omics types enriched in disease-related pathways and their associated interactions (such as protein-protein, protein-transcript, protein-metabolite, transcript-metabolite, and lipid-lipid interactions) that are unique to mild, moderate, and severe COVID-19 disease states. These biosignatures include molecular features that underlie the observed clinical heterogeneity of COVID-19 and emphasize the need for disease-phase-specific treatment strategies. The approach implemented here can be used to find associations between transcripts, proteins, lipids, and metabolites in other diseases.
Collapse
Affiliation(s)
- Francis E. Agamah
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Thomas H. A. Ederveen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - Michelle Skelton
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Darren P. Martin
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Emile R. Chimusa
- Department of Applied Science, Faculty of Health and Life Sciences, Northumbria University, Newcastle, United Kingdom
| | - Peter A. C. ’t Hoen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| |
Collapse
|
21
|
Shouman S, El-Kholy N, Hussien AE, El-Derby AM, Magdy S, Abou-Shanab AM, Elmehrath AO, Abdelwaly A, Helal M, El-Badri N. SARS-CoV-2-associated lymphopenia: possible mechanisms and the role of CD147. Cell Commun Signal 2024; 22:349. [PMID: 38965547 PMCID: PMC11223399 DOI: 10.1186/s12964-024-01718-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 06/15/2024] [Indexed: 07/06/2024] Open
Abstract
T lymphocytes play a primary role in the adaptive antiviral immunity. Both lymphocytosis and lymphopenia were found to be associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While lymphocytosis indicates an active anti-viral response, lymphopenia is a sign of poor prognosis. T-cells, in essence, rarely express ACE2 receptors, making the cause of cell depletion enigmatic. Moreover, emerging strains posed an immunological challenge, potentially alarming for the next pandemic. Herein, we review how possible indirect and direct key mechanisms could contribute to SARS-CoV-2-associated-lymphopenia. The fundamental mechanism is the inflammatory cytokine storm elicited by viral infection, which alters the host cell metabolism into a more acidic state. This "hyperlactic acidemia" together with the cytokine storm suppresses T-cell proliferation and triggers intrinsic/extrinsic apoptosis. SARS-CoV-2 infection also results in a shift from steady-state hematopoiesis to stress hematopoiesis. Even with low ACE2 expression, the presence of cholesterol-rich lipid rafts on activated T-cells may enhance viral entry and syncytia formation. Finally, direct viral infection of lymphocytes may indicate the participation of other receptors or auxiliary proteins on T-cells, that can work alone or in concert with other mechanisms. Therefore, we address the role of CD147-a novel route-for SARS-CoV-2 and its new variants. CD147 is not only expressed on T-cells, but it also interacts with other co-partners to orchestrate various biological processes. Given these features, CD147 is an appealing candidate for viral pathogenicity. Understanding the molecular and cellular mechanisms behind SARS-CoV-2-associated-lymphopenia will aid in the discovery of potential therapeutic targets to improve the resilience of our immune system against this rapidly evolving virus.
Collapse
Affiliation(s)
- Shaimaa Shouman
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Nada El-Kholy
- Department of Drug Discovery, H. Lee Moffit Cancer Center& Research Institute, Tampa, FL, 33612, USA
- Cancer Chemical Biology Ph.D. Program, University of South Florida, Tampa, FL, 33620, USA
| | - Alaa E Hussien
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Azza M El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Shireen Magdy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Ahmed M Abou-Shanab
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | | | - Ahmad Abdelwaly
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
- Institute for Computational Molecular Science, Department of Chemistry, Temple University, Philadelphia, PA, 19122, USA
| | - Mohamed Helal
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt.
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt.
| |
Collapse
|
22
|
Q.B. Alenzi F. Survivin: A key apoptosis inhibitor in COVID-19 infection and its implication for treatment protocol. Saudi J Biol Sci 2024; 31:104021. [PMID: 38831893 PMCID: PMC11145386 DOI: 10.1016/j.sjbs.2024.104021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 06/05/2024] Open
Abstract
While the relationship between cellular apoptosis and proliferation rates in COVID patients remains underexplored in existing literature, various viruses are known to impact these fundamental process to modulate response to infection. This paper aims to assess apoptosis and proliferation rates in individuals recently infected with Coronavirus, both before and after vaccination, comparing them with healthy controls. Peripheral blood cells from newly diagnosed COVID-19 patients revealed a significant increase in proliferation and apoptosis levels in fresh lymphocytes and granulocytes compared to healthy donors. Notably, as none of the patients were under corticosteroid therapy or cytotoxic drugs, the study underscores the critical role of white blood (WBC) apoptosis in viral pathogenesis, potentially contributing significantly to COVID-19's pathogenicity. Elevated levels of soluble Fas ligand (FaSL) and the pro-inflatmmatory cytokine IL-38 were identified in COVID-19 patients, indicating potential immune dysregulation. Furthermore, individual who received the vaccine or recovered from COVID-19 exhibited higher survivin rates, suggesting a protective role for survivin in migitating lung damage. These findings suggest the prospect of developing a strategy to prevent WBC apoptosis, offering potential benefits in averting lymphopenia associated with severe COVID-19 ouctomes.
Collapse
Affiliation(s)
- Faris Q.B. Alenzi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
23
|
Albert MC, Uranga-Murillo I, Arias M, De Miguel D, Peña N, Montinaro A, Varanda AB, Theobald SJ, Areso I, Saggau J, Koch M, Liccardi G, Peltzer N, Rybniker J, Hurtado-Guerrero R, Merino P, Monzón M, Badiola JJ, Reindl-Schwaighofer R, Sanz-Pamplona R, Cebollada-Solanas A, Megyesfalvi Z, Dome B, Secrier M, Hartmann B, Bergmann M, Pardo J, Walczak H. Identification of FasL as a crucial host factor driving COVID-19 pathology and lethality. Cell Death Differ 2024; 31:544-557. [PMID: 38514848 PMCID: PMC11093991 DOI: 10.1038/s41418-024-01278-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
The dysregulated immune response and inflammation resulting in severe COVID-19 are still incompletely understood. Having recently determined that aberrant death-ligand-induced cell death can cause lethal inflammation, we hypothesized that this process might also cause or contribute to inflammatory disease and lung failure following SARS-CoV-2 infection. To test this hypothesis, we developed a novel mouse-adapted SARS-CoV-2 model (MA20) that recapitulates key pathological features of COVID-19. Concomitantly with occurrence of cell death and inflammation, FasL expression was significantly increased on inflammatory monocytic macrophages and NK cells in the lungs of MA20-infected mice. Importantly, therapeutic FasL inhibition markedly increased survival of both, young and old MA20-infected mice coincident with substantially reduced cell death and inflammation in their lungs. Intriguingly, FasL was also increased in the bronchoalveolar lavage fluid of critically-ill COVID-19 patients. Together, these results identify FasL as a crucial host factor driving the immuno-pathology that underlies COVID-19 severity and lethality, and imply that patients with severe COVID-19 may significantly benefit from therapeutic inhibition of FasL.
Collapse
Affiliation(s)
- Marie-Christine Albert
- Cell death, inflammation and immunity laboratory, CECAD Cluster of Excellence, University of Cologne, Cologne, 50931, Germany
- Cell death, inflammation and immunity laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, 50931, Germany
| | - Iratxe Uranga-Murillo
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, 28029, Spain
- Aragón Health Research Institute (IIS Aragón), San Juan Bosco 13, Zaragoza, 50009, Spain
- Department of Microbiology, Paediatrics, Radiology and Preventive Medicine and Public Health, University of Zaragoza, Zaragoza, 50009, Spain
| | - Maykel Arias
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, 28029, Spain
- Aragón Health Research Institute (IIS Aragón), San Juan Bosco 13, Zaragoza, 50009, Spain
- Department of Microbiology, Paediatrics, Radiology and Preventive Medicine and Public Health, University of Zaragoza, Zaragoza, 50009, Spain
| | - Diego De Miguel
- Aragón Health Research Institute (IIS Aragón), San Juan Bosco 13, Zaragoza, 50009, Spain
| | - Natacha Peña
- Aragón Health Research Institute (IIS Aragón), San Juan Bosco 13, Zaragoza, 50009, Spain
| | - Antonella Montinaro
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, WC1E 6DD, UK
| | - Ana Beatriz Varanda
- Cell death, inflammation and immunity laboratory, CECAD Cluster of Excellence, University of Cologne, Cologne, 50931, Germany
- Cell death, inflammation and immunity laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, 50931, Germany
| | - Sebastian J Theobald
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, 50931, Germany
- Faculty of Medicine and University Hospital of Cologne, Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, 50931, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, 50931, Germany
| | - Itziar Areso
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, WC1E 6DD, UK
| | - Julia Saggau
- Cell death, inflammation and immunity laboratory, CECAD Cluster of Excellence, University of Cologne, Cologne, 50931, Germany
- Cell death, inflammation and immunity laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, 50931, Germany
- Genome instability, inflammation and cell death laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, 50931, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, 50931, Germany
| | - Manuel Koch
- Institue for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, Cologne, 50931, Germany
| | - Gianmaria Liccardi
- Genome instability, inflammation and cell death laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, 50931, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, 50931, Germany
| | - Nieves Peltzer
- Cell death, inflammation and immunity laboratory, CECAD Cluster of Excellence, University of Cologne, Cologne, 50931, Germany
- Faculty of Medicine and University Hospital of Cologne, Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, 50931, Germany
- Department of Translational Genomics, University of Cologne, Cologne, 50931, Germany
| | - Jan Rybniker
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, 50931, Germany
- Faculty of Medicine and University Hospital of Cologne, Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, 50931, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, 50931, Germany
| | - Ramón Hurtado-Guerrero
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), University of Zaragoza, Zaragoza, 50018, Spain
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, 2200, Denmark
- Fundación ARAID, Zaragoza, 50018, Spain
| | - Pedro Merino
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), University of Zaragoza, Zaragoza, 50018, Spain
| | - Marta Monzón
- Research Centre for Encephalopaties and Transmissible Emerging Diseases, Institute for Health Research Aragón (IIS), University of Zaragoza, Zaragoza, 50013, Spain
- Department of Human Anatomy and Histology, University of Zaragoza, Zaragoza, 50009, Spain
| | - Juan J Badiola
- Research Centre for Encephalopaties and Transmissible Emerging Diseases, Institute for Health Research Aragón (IIS), University of Zaragoza, Zaragoza, 50013, Spain
| | | | - Rebeca Sanz-Pamplona
- Aragón Health Research Institute (IIS Aragón), San Juan Bosco 13, Zaragoza, 50009, Spain
- Fundación ARAID, Zaragoza, 50018, Spain
- CIBER de Epidemiología y Salud Pública, Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Alberto Cebollada-Solanas
- Aragon Biomedical Research Center (CIBA), Instituto Aragonés de Ciencias de la Salud (IACS), Unidad de Biocomputación, Zaragoza, 50018, Spain
| | - Zsolt Megyesfalvi
- Deparment of Thoracic Surgery, Medical University of Vienna, Vienna, 1090, Austria
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, 1122, Hungary
- National Koranyi Institute of Pulmonology, Budapest, 1121, Hungary
| | - Balazs Dome
- Deparment of Thoracic Surgery, Medical University of Vienna, Vienna, 1090, Austria
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, 1122, Hungary
- National Koranyi Institute of Pulmonology, Budapest, 1121, Hungary
- Department of Translational Medicine, Lund University, Lund, SE-22100, Sweden
| | - Maria Secrier
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, United Kingdom
| | - Boris Hartmann
- Virology Group, Institute for Veterinary Disease Control at AGES, Moedling, 2340, Austria
| | - Michael Bergmann
- Div. of Visceral Surgery, Dept. of General Surgery, Comprehensive Cancer Centre, Medical University of Vienna, Vienna, 1090, Austria
| | - Julián Pardo
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, 28029, Spain
- Aragón Health Research Institute (IIS Aragón), San Juan Bosco 13, Zaragoza, 50009, Spain
- Department of Microbiology, Paediatrics, Radiology and Preventive Medicine and Public Health, University of Zaragoza, Zaragoza, 50009, Spain
| | - Henning Walczak
- Cell death, inflammation and immunity laboratory, CECAD Cluster of Excellence, University of Cologne, Cologne, 50931, Germany.
- Cell death, inflammation and immunity laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, 50931, Germany.
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, WC1E 6DD, UK.
| |
Collapse
|
24
|
Luo Y, Zhang Z, Ren J, Dou C, Wen J, Yang Y, Li X, Yan Z, Han Y. SARS-Cov-2 spike induces intestinal barrier dysfunction through the interaction between CEACAM5 and Galectin-9. Front Immunol 2024; 15:1303356. [PMID: 38686388 PMCID: PMC11056506 DOI: 10.3389/fimmu.2024.1303356] [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: 09/27/2023] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
Background Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), as a typical tumor marker, has been found to exert immunomodulatory effects in many diseases. We previously reported the clinical and molecular evidences supporting that SARS-Cov-2 infected the gastrointestinal (GI) tract and found a reduction of CEACAM5 in COVID-19 patients' feces which associated with gut dysbiosis. Yet the role of CEACAM5 in GI infection is ill-defined. Methods Mice models were established through intraperitoneally injecting with recombinant viral spike-Fc to mimic the intestinal inflammation. We collected duodenum, jejunum, ileum and colon samples after 6h, 2 days, 4 days and 7 days of spike-Fc or control-Fc injection to perform proteomic analysis. Blood was collected from healthy donors and peripheral blood mononuclear cells (PBMC) were separated by density gradient centrifugation, then CD4+ T cells were isolated with magnetic beads and co-cultured with Caco-2 cells. Results In addition to intestinal CEACAM5, the expression of tight junction and the percent of CD4+ T lymphocytes were significantly decreased in spike-Fc group compared to control (p < 0.05), accompanied with increased level of inflammatory factors. The KEGG analysis revealed differentially expressed proteins were mainly enriched in the coronavirus disease (COVID-19), tight junction, focal adhesion, adherens junction and PI3K-Akt signaling pathway. Protein-protein interaction (PPI) network analysis identified the interaction between CEACAM5 and Galectin-9 that was also verified by molecular docking and co-IP assay. We further confirmed a reduction of CEACAM5 in SARS-CoV-2 spike stimulated enterocytes could promote the expression of Galectin-9 protein in CD4+T cells. Then it gave rise to the increasing release of inflammatory factors and increased apoptosis of CD4+T cells by inhibition of PI3K/AKT/mTOR pathway. Ultimately intestinal barrier dysfunction happened. Conclusion Our results indicated that CEACAM5 overexpression and Galectin-9 knockdown played a protective role in intestinal barrier injury upon spike-Fc stimulation. Collectively, our findings identified firstly that SARS-CoV-2 spike induced intestinal barrier dysfunction through the interaction between CEACAM5 and Galectin-9. The result provides potential therapeutic targets in intestinal barrier dysfunction for treating severe COVID patients.
Collapse
Affiliation(s)
- Yingshu Luo
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Zhenling Zhang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jiangnan Ren
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Chunxu Dou
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jiancheng Wen
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yang Yang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Xiaofeng Li
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Zhixiang Yan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yanzhi Han
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| |
Collapse
|
25
|
Campagna R, Dominelli F, Zingaropoli MA, Ciurluini F, Grilli G, Amoroso A, De Domenico A, Amatore D, Lia MS, Cortesi E, Picone V, Mastroianni CM, Ciardi MR, De Santis R, Lista F, Antonelli G, Turriziani O. COVID-19 vaccination in cancer patients: Immune responses one year after the third dose. Vaccine 2024; 42:2687-2694. [PMID: 38499458 DOI: 10.1016/j.vaccine.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/20/2024]
Abstract
Cancer patients (CPs), being immunosuppressed due to the treatment received or to the disease itself, are more susceptible to infections and their potential complications, showing therefore an increased risk of developing severe COVID-19 compared to the general population. We evaluated the immune responses to anti-SARS-CoV-2 vaccination in patients with solid tumors one year after the administration of the third dose and the effect of cancer treatment on vaccine immunogenicity was assessed. Healthy donors (HDs) were enrolled. Binding and neutralizing antibody (Ab) titers were evaluated using chemiluminescence immunoassay (CLIA) and Plaque Reduction Neutralization Test (PRNT) respectively. T-cell response was analyzed using multiparametric flow cytometry. CPs who were administered three vaccine doses showed lower Ab titers than CPs with four doses and HDs. Overall, a lower cell-mediated response was found in CPs, with a predominance of monofunctional T-cells producing TNFα. Lower Ab titers and a weaker T-cell response were observed in CPs without prior SARS-CoV-2 infection when compared to those with a previous infection. While no differences in the humoral response were found comparing immunotherapy and non-immunotherapy patients, a stronger T-cell response in CPs treated with immunotherapy was observed. Our results emphasize the need of booster doses in cancer patients to achieve a level of protection similar to that observed in healthy donors and underlines the importance of considering the treatment received to reach a proper immune response.
Collapse
Affiliation(s)
- Roberta Campagna
- Department of Molecular Medicine Sapienza University of Rome, Viale dell'Università, 33, 000185 Rome, Italy.
| | - Federica Dominelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy.
| | - Maria Antonella Zingaropoli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy.
| | - Fabio Ciurluini
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185 Rome, Italy.
| | - Giorgia Grilli
- Defence Institute for Biomedical Sciences, 00184 Rome, Italy.
| | | | | | | | | | - Enrico Cortesi
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185 Rome, Italy.
| | - Vincenzo Picone
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185 Rome, Italy.
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy.
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy.
| | - Riccardo De Santis
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy; Defence Institute for Biomedical Sciences, 00184 Rome, Italy.
| | - Florigio Lista
- Defence Institute for Biomedical Sciences, 00184 Rome, Italy.
| | - Guido Antonelli
- Department of Molecular Medicine Sapienza University of Rome, Viale dell'Università, 33, 000185 Rome, Italy.
| | - Ombretta Turriziani
- Department of Molecular Medicine Sapienza University of Rome, Viale dell'Università, 33, 000185 Rome, Italy.
| |
Collapse
|
26
|
Purwono PB, Vacharathit V, Manopwisedjaroen S, Ludowyke N, Suksatu A, Thitithanyanont A. Infection kinetics, syncytia formation, and inflammatory biomarkers as predictive indicators for the pathogenicity of SARS-CoV-2 Variants of Concern in Calu-3 cells. PLoS One 2024; 19:e0301330. [PMID: 38568894 PMCID: PMC10990222 DOI: 10.1371/journal.pone.0301330] [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: 07/02/2023] [Accepted: 03/13/2024] [Indexed: 04/05/2024] Open
Abstract
The ongoing COVID-19 pandemic has led to the emergence of new SARS-CoV-2 variants as a result of continued host-virus interaction and viral genome mutations. These variants have been associated with varying levels of transmissibility and disease severity. We investigated the phenotypic profiles of six SARS-CoV-2 variants (WT, D614G, Alpha, Beta, Delta, and Omicron) in Calu-3 cells, a human lung epithelial cell line. In our model demonstrated that all variants, except for Omicron, had higher efficiency in virus entry compared to the wild-type. The Delta variant had the greatest phenotypic advantage in terms of early infection kinetics and marked syncytia formation, which could facilitate cell-to-cell spreading, while the Omicron variant displayed slower replication and fewer syncytia formation. We also identified the Delta variant as the strongest inducer of inflammatory biomarkers, including pro-inflammatory cytokines/chemokines (IP-10/CXCL10, TNF-α, and IL-6), anti-inflammatory cytokine (IL-1RA), and growth factors (FGF-2 and VEGF-A), while these inflammatory mediators were not significantly elevated with Omicron infection. These findings are consistent with the observations that there was a generally more pronounced inflammatory response and angiogenesis activity within the lungs of COVID-19 patients as well as more severe symptoms and higher mortality rate during the Delta wave, as compared to less severe symptoms and lower mortality observed during the current Omicron wave in Thailand. Our findings suggest that early infectivity kinetics, enhanced syncytia formation, and specific inflammatory mediator production may serve as predictive indicators for the virulence potential of future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Priyo Budi Purwono
- Faculty of Science, Department of Microbiology, Mahidol University, Bangkok, Thailand
- Faculty of Medicine, Department of Microbiology, Universitas Airlangga, Surabaya, Indonesia
| | - Vimvara Vacharathit
- Faculty of Science, Department of Microbiology, Mahidol University, Bangkok, Thailand
- Faculty of Science, Systems Biology of Diseases Research Unit, Mahidol University, Bangkok, Thailand
| | | | - Natali Ludowyke
- Faculty of Science, Department of Microbiology, Mahidol University, Bangkok, Thailand
| | - Ampa Suksatu
- Faculty of Science, Department of Microbiology, Mahidol University, Bangkok, Thailand
| | - Arunee Thitithanyanont
- Faculty of Science, Department of Microbiology, Mahidol University, Bangkok, Thailand
- Faculty of Science, Department of Microbiology, Pornchai Matangkasombut Center for Microbial Genomics, Mahidol University, Bangkok, Thailand
| |
Collapse
|
27
|
Kundura L, Cezar R, Ballongue E, André S, Michel M, Mettling C, Lozano C, Vincent T, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Loubet P, Sotto A, Tran TA, Estaquier J, Corbeau P. Low Percentage of Perforin-Expressing NK Cells during Severe SARS-CoV-2 Infection: Consumption Rather than Primary Deficiency. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1105-1112. [PMID: 38345346 DOI: 10.4049/jimmunol.2300359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 12/05/2023] [Indexed: 03/20/2024]
Abstract
Genetic defects in the ability to deliver effective perforin have been reported in patients with hemophagocytic lymphohistiocytosis. We tested the hypothesis that a primary perforin deficiency might also be causal in severe SARS-CoV-2 infection. We recruited 54 volunteers confirmed as being SARS-CoV-2-infected by RT-PCR and admitted to intensive care units or non-intensive care units and age- and sex-matched healthy controls. Compared with healthy controls, the percentage of perforin-expressing CD3-CD56+ NK cells quantified by flow cytometry was low in COVID-19 patients (69.9 ± 17.7 versus 78.6 ± 14.6%, p = 0.026). There was no correlation between the proportions of perforin-positive NK cells and T8 lymphocytes. Moreover, the frequency of NK cells producing perforin was neither linked to disease severity nor predictive of death. Although IL-6 is known to downregulate perforin production in NK cells, we did not find any link between perforin expression and IL-6 plasma level. However, we unveiled a negative correlation between the degranulation marker CD107a and perforin expression in NK cells (r = -0.488, p = 10-4). PRF1 gene expression and the frequency of NK cells harboring perforin were normal in patients 1 y after acute SARS-CoV-2 infection. A primary perforin defect does not seem to be a driver of COVID-19 because NK perforin expression is 1) linked neither to T8 perforin expression nor to disease severity, 2) inversely correlated with NK degranulation, and 3) normalized at distance from acute infection. Thus, the cause of low frequency of perforin-positive NK cells appears, rather, to be consumption.
Collapse
Affiliation(s)
- Lucy Kundura
- Institute of Human Genetics, UMR9002, CNRS and Montpellier University, Montpellier, France
| | - Renaud Cezar
- Immunology Department, Nîmes University Hospital, Nîmes, France
| | - Emma Ballongue
- Institute of Human Genetics, UMR9002, CNRS and Montpellier University, Montpellier, France
| | - Sonia André
- INSERM U1124, Université de Paris, Paris, France
| | - Moïse Michel
- Immunology Department, Nîmes University Hospital, Nîmes, France
| | - Clément Mettling
- Institute of Human Genetics, UMR9002, CNRS and Montpellier University, Montpellier, France
| | - Claire Lozano
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Thierry Vincent
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Laurent Muller
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Jean-Yves Lefrant
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Claire Roger
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Pierre-Géraud Claret
- Medical and Surgical Emergency Department, Nîmes University Hospital, Nîmes, France
| | - Sandra Duvnjak
- Gerontology Department, Nîmes University Hospital, Nîmes, France
| | - Paul Loubet
- *Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Albert Sotto
- *Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Tu-Anh Tran
- Pediatrics Department, Nîmes University Hospital, Nîmes, France
| | - Jérôme Estaquier
- INSERM U1124, Université de Paris, Paris, France
- Laval University Research Center; Quebec City, Quebec, Canada
| | - Pierre Corbeau
- Institute of Human Genetics, UMR9002, CNRS and Montpellier University, Montpellier, France
- Immunology Department, Nîmes University Hospital, Nîmes, France
| |
Collapse
|
28
|
Chen WC, Hu SY, Cheng CM, Shen CF, Chuang HY, Ker CR, Sun DJ, Shen CJ. TRAIL and IP-10 dynamics in pregnant women post COVID-19 vaccination: associations with neutralizing antibody potency. Front Cell Infect Microbiol 2024; 14:1358967. [PMID: 38572318 PMCID: PMC10987851 DOI: 10.3389/fcimb.2024.1358967] [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: 12/20/2023] [Accepted: 03/07/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction The aim of this study is to investigate changes in TNF-related apoptosis-inducing ligand (TRAIL) and gamma interferon-induced protein 10 (IP-10) after COVID-19 vaccination in pregnant women and to explore their association with neutralizing antibody (Nab) inhibition. Methods The study evaluated 93 pregnant women who had previously received two (n=21), three (n=55) or four (n=17) doses of COVID-19 vaccine. Also we evaluated maternal blood samples that were collected during childbirth. The levels of TRAIL, IP-10 and Nab inhibition were measured using enzyme-linked immunosorbent assays (ELISA). Results and discussion Our study revealed four-dose group resulted in lower TRAIL levels when compared to the two-dose and three-dose groups (4.78 vs. 16.07 vs. 21.61 pg/ml, p = 0.014). The two-dose group had reduced IP-10 levels than the three-dose cohort (111.49 vs. 147.89 pg/ml, p=0.013), with no significant variation compared to the four-dose group. In addition, the four-dose group showed stronger Nab inhibition against specific strains (BA.2 and BA.5) than the three-dose group. A positive correlation was observed between TRAIL and IP-10 in the two-dose group, while this relationship was not found in other dose groups or between TRAIL/IP-10 and Nab inhibition. As the doses of the COVID-19 vaccine increase, the levels of TRAIL and IP-10 generally increase, only by the fourth dose, the group previously vaccinated with AZD1222 showed lower TRAIL but higher IP-10. Despite these changes, more doses of the vaccine consistently reinforced Nab inhibition, apparently without any relation to TRAIL and IP-10 levels. The variation may indicate the induction of immunological memory in vaccinated mothers, which justifies further research in the future.
Collapse
Affiliation(s)
- Wei-Chun Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Obstetrics and Gynecology, New Taipei City Municipal Tucheng Hospital, New Taipei City, Taiwan
- International Intercollegiate Ph.D. Program, National Tsing Hua University, Hsinchu, Taiwan
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shu-Yu Hu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Fen Shen
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Yu Chuang
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin-Ru Ker
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Der-Ji Sun
- Department of Obstetrics and Gynecology, Pojen Hospital, Kaohsiung, Taiwan
| | - Ching-Ju Shen
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
29
|
Michels EHA, Appelman B, de Brabander J, van Amstel RBE, van Linge CCA, Chouchane O, Reijnders TDY, Schuurman AR, Sulzer TAL, Klarenbeek AM, Douma RA, Bos LDJ, Wiersinga WJ, Peters-Sengers H, van der Poll T. Host Response Changes and Their Association with Mortality in COVID-19 Patients with Lymphopenia. Am J Respir Crit Care Med 2024; 209:402-416. [PMID: 37948687 PMCID: PMC10878379 DOI: 10.1164/rccm.202305-0890oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/09/2023] [Indexed: 11/12/2023] Open
Abstract
Rationale: Lymphopenia in coronavirus disease (COVID-19) is associated with increased mortality. Objectives: To explore the association between lymphopenia, host response aberrations, and mortality in patients with lymphopenic COVID-19. Methods: We determined 43 plasma biomarkers reflective of four pathophysiological domains: endothelial cell and coagulation activation, inflammation and organ damage, cytokine release, and chemokine release. We explored if decreased concentrations of lymphocyte-derived proteins in patients with lymphopenia were associated with an increase in mortality. We sought to identify host response phenotypes in patients with lymphopenia by cluster analysis of plasma biomarkers. Measurements and Main Results: A total of 439 general ward patients with COVID-19 were stratified by baseline lymphocyte counts: normal (>1.0 × 109/L; n = 167), mild lymphopenia (>0.5 to ⩽1.0 × 109/L; n = 194), and severe lymphopenia (⩽0.5 × 109/L; n = 78). Lymphopenia was associated with alterations in each host response domain. Lymphopenia was associated with increased mortality. Moreover, in patients with lymphopenia (n = 272), decreased concentrations of several lymphocyte-derived proteins (e.g., CCL5, IL-4, IL-13, IL-17A) were associated with an increase in mortality (at P < 0.01 or stronger significance levels). A cluster analysis revealed three host response phenotypes in patients with lymphopenia: "hyporesponsive" (23.2%), "hypercytokinemic" (36.4%), and "inflammatory-injurious" (40.4%), with substantially differing mortality rates of 9.5%, 5.1%, and 26.4%, respectively. A 10-biomarker model accurately predicted these host response phenotypes in an external cohort with similar mortality distribution. The inflammatory-injurious phenotype showed a remarkable combination of relatively high inflammation and organ damage markers with high antiinflammatory cytokine levels yet low proinflammatory cytokine levels. Conclusions: Lymphopenia in COVID-19 signifies a heterogenous group of patients with distinct host response features. Specific host responses contribute to lymphopenia-associated mortality in COVID-19, including reduced CCL5 levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Renée A. Douma
- Department of Internal Medicine, Flevo Hospital, Almere, the Netherlands; and
| | | | - W. Joost Wiersinga
- Center for Experimental and Molecular Medicine
- Division of Infectious Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Hessel Peters-Sengers
- Center for Experimental and Molecular Medicine
- Department of Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine
- Division of Infectious Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
30
|
Vujkovic A, Ha M, de Block T, van Petersen L, Brosius I, Theunissen C, van Ierssel SH, Bartholomeus E, Adriaensen W, Vanham G, Elias G, Van Damme P, Van Tendeloo V, Beutels P, van Frankenhuijsen M, Vlieghe E, Ogunjimi B, Laukens K, Meysman P, Vercauteren K. Diagnosing Viral Infections Through T-Cell Receptor Sequencing of Activated CD8+ T Cells. J Infect Dis 2024; 229:507-516. [PMID: 37787611 PMCID: PMC10873181 DOI: 10.1093/infdis/jiad430] [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: 05/11/2023] [Revised: 08/26/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023] Open
Abstract
T-cell-based diagnostic tools identify pathogen exposure but lack differentiation between recent and historical exposures in acute infectious diseases. Here, T-cell receptor (TCR) RNA sequencing was performed on HLA-DR+/CD38+CD8+ T-cell subsets of hospitalized coronavirus disease 2019 (COVID-19) patients (n = 30) and healthy controls (n = 30; 10 of whom had previously been exposed to severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2]). CDR3α and CDR3β TCR regions were clustered separately before epitope specificity annotation using a database of SARS-CoV-2-associated CDR3α and CDR3β sequences corresponding to >1000 SARS-CoV-2 epitopes. The depth of the SARS-CoV-2-associated CDR3α/β sequences differentiated COVID-19 patients from the healthy controls with a receiver operating characteristic area under the curve of 0.84 ± 0.10. Hence, annotating TCR sequences of activated CD8+ T cells can be used to diagnose an acute viral infection and discriminate it from historical exposure. In essence, this work presents a new paradigm for applying the T-cell repertoire to accomplish TCR-based diagnostics.
Collapse
Affiliation(s)
- Alexandra Vujkovic
- Clinical Virology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
| | - My Ha
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Antwerp, Belgium
- Centre for Health Economics Research and Modeling Infectious Diseases (CHERMID), University of Antwerp, Belgium
- Vaccine and Infectious Disease Institute, University of Antwerp, Belgium
| | - Tessa de Block
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Lida van Petersen
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Isabel Brosius
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Caroline Theunissen
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Sabrina H van Ierssel
- Department of General Internal Medicine, Infectious Diseases and Tropical Medicine, University Hospital Antwerp, Belgium
| | - Esther Bartholomeus
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Antwerp, Belgium
| | - Wim Adriaensen
- Clinical Immunology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Guido Vanham
- Biomedical Department, Institute of Tropical Medicine, Antwerp, Belgium
| | - George Elias
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, University of Antwerp, Belgium
| | - Pierre Van Damme
- Vaccine and Infectious Disease Institute, University of Antwerp, Belgium
| | - Viggo Van Tendeloo
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, University of Antwerp, Belgium
| | - Philippe Beutels
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Antwerp, Belgium
- Centre for Health Economics Research and Modeling Infectious Diseases (CHERMID), University of Antwerp, Belgium
| | | | - Erika Vlieghe
- Department of General Internal Medicine, Infectious Diseases and Tropical Medicine, University Hospital Antwerp, Belgium
| | - Benson Ogunjimi
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Antwerp, Belgium
- Centre for Health Economics Research and Modeling Infectious Diseases (CHERMID), University of Antwerp, Belgium
- Vaccine and Infectious Disease Institute, University of Antwerp, Belgium
- Department of Paediatrics, Antwerp University Hospital, Antwerp, Belgium
| | - Kris Laukens
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
| | - Pieter Meysman
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
| | - Koen Vercauteren
- Clinical Virology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| |
Collapse
|
31
|
Harriott NC, Ryan AL. Proteomic profiling identifies biomarkers of COVID-19 severity. Heliyon 2024; 10:e23320. [PMID: 38163173 PMCID: PMC10755324 DOI: 10.1016/j.heliyon.2023.e23320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
SARS-CoV-2 infection remains a major public health concern, particularly for the aged and those individuals with co-morbidities at risk for developing severe COVID-19. Understanding the pathogenesis and biomarkers associated with responses to SARS-CoV-2 infection remain critical components in developing effective therapeutic approaches, especially in cases of severe and long-COVID-19. In this study blood plasma protein expression was compared in subjects with mild, moderate, and severe COVID-19 disease. Evaluation of an inflammatory protein panel confirms upregulation of proteins including TNFβ, IL-6, IL-8, IL-12, already associated with severe cytokine storm and progression to severe COVID-19. Importantly, we identify several proteins not yet associated with COVID-19 disease, including mesothelin (MSLN), that are expressed at significantly higher levels in severe COVID-19 subjects. In addition, we find a subset of markers associated with T-cell and dendritic cell responses to viral infection that are significantly higher in mild cases and decrease in expression as severity of COVID-19 increases, suggesting that an immediate and effective activation of T-cells is critical in modulating disease progression. Together, our findings identify new targets for further investigation as therapeutic approaches for the treatment of SARS-CoV-2 infection and prevention of complications of severe COVID-19.
Collapse
Affiliation(s)
- Noa C. Harriott
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City IA 52240, USA
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City IA 52240, USA
| |
Collapse
|
32
|
Safont G, Villar-Hernández R, Smalchuk D, Stojanovic Z, Marín A, Lacoma A, Pérez-Cano C, López-Martínez A, Molina-Moya B, Solis AJ, Arméstar F, Matllo J, Díaz-Fernández S, Romero I, Casas I, Strecker K, Preyer R, Rosell A, Latorre I, Domínguez J. Measurement of IFN-γ and IL-2 for the assessment of the cellular immunity against SARS-CoV-2. Sci Rep 2024; 14:1137. [PMID: 38212416 PMCID: PMC10784529 DOI: 10.1038/s41598-024-51505-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024] Open
Abstract
The study of specific T-cell responses against SARS-CoV-2 is important for understanding long-term immunity and infection management. The aim of this study was to assess the dual IFN-γ and IL-2 detection, using a SARS-CoV-2 specific fluorescence ELISPOT, in patients undergoing acute disease, during convalescence, and after vaccination. We also evaluated humoral response and compared with T-cells with the aim of correlating both types of responses, and increase the number of specific response detection. Blood samples were drawn from acute COVID-19 patients and convalescent individuals classified according to disease severity; and from unvaccinated and vaccinated uninfected individuals. IgGs against Spike and nucleocapsid, IgMs against nucleocapsid, and neutralizing antibodies were also analyzed. Our results show that IFN-γ in combination with IL-2 increases response detection in acute and convalescent individuals (p = 0.023). In addition, IFN-γ detection can be a useful biomarker for monitoring severe acute patients, as our results indicate that those individuals with a poor outcome have lower levels of this cytokine. In some cases, the lack of cellular immunity is compensated by antibodies, confirming the role of both types of immune responses in infection, and confirming that their dual detection can increase the number of specific response detections. In summary, IFN-γ/IL-2 dual detection is promising for characterizing and assessing the immunization status, and helping in the patient management.
Collapse
Affiliation(s)
- Guillem Safont
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Raquel Villar-Hernández
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- Genome Identification Diagnostics GmbH (GenID), Straßberg, Germany
| | - Daria Smalchuk
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- Odesa I. I. Mechnykov National University, Odesa, Ukraine
| | - Zoran Stojanovic
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Alicia Marín
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Alicia Lacoma
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Pérez-Cano
- Basic Unit for the Prevention of Occupational Risks (UBP), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Anabel López-Martínez
- Basic Unit for the Prevention of Occupational Risks (UBP), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Bárbara Molina-Moya
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alan Jhunior Solis
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Fernando Arméstar
- Intensive Care Medicine Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
| | - Joan Matllo
- Basic Unit for the Prevention of Occupational Risks (UBP), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Sergio Díaz-Fernández
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Iris Romero
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Irma Casas
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Preventive Medicine Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Kevin Strecker
- Genome Identification Diagnostics GmbH (GenID), Straßberg, Germany
| | - Rosemarie Preyer
- Genome Identification Diagnostics GmbH (GenID), Straßberg, Germany
| | - Antoni Rosell
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Irene Latorre
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jose Domínguez
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain.
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
33
|
Cezar R, Kundura L, André S, Lozano C, Vincent T, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Loubet P, Sotto A, Tran TA, Estaquier J, Corbeau P. T4 apoptosis in the acute phase of SARS-CoV-2 infection predicts long COVID. Front Immunol 2024; 14:1335352. [PMID: 38235145 PMCID: PMC10791767 DOI: 10.3389/fimmu.2023.1335352] [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: 11/08/2023] [Accepted: 12/07/2023] [Indexed: 01/19/2024] Open
Abstract
Background As about 10% of patients with COVID-19 present sequelae, it is important to better understand the physiopathology of so-called long COVID. Method To this aim, we recruited 29 patients hospitalized for SARS-CoV-2 infection and, by Luminex®, quantified 19 soluble factors in their plasma and in the supernatant of their peripheral blood mononuclear cells, including inflammatory and anti-inflammatory cytokines and chemokines, Th1/Th2/Th17 cytokines, and endothelium activation markers. We also measured their T4, T8 and NK differentiation, activation, exhaustion and senescence, T cell apoptosis, and monocyte subpopulations by flow cytometry. We compared these markers between participants who developed long COVID or not one year later. Results None of these markers was predictive for sequelae, except programmed T4 cell death. T4 lymphocytes from participants who later presented long COVID were more apoptotic in culture than those of sequelae-free participants at Month 12 (36.9 ± 14.7 vs. 24.2 ± 9.0%, p = 0.016). Conclusions Our observation raises the hypothesis that T4 cell death during the acute phase of SARS-CoV-2 infection might pave the way for long COVID. Mechanistically, T4 lymphopenia might favor phenomena that could cause sequelae, including SARS-CoV-2 persistence, reactivation of other viruses, autoimmunity and immune dysregulation. In this scenario, inhibiting T cell apoptosis, for instance, by caspase inhibitors, could prevent long COVID.
Collapse
Affiliation(s)
- Renaud Cezar
- Immunology Department, Nîmes University Hospital, Nîmes, France
| | - Lucy Kundura
- Institute of Human Genetics, UMR9002, Centre National de la Recherche Scientifique (CNRS) and Montpellier University, Montpellier, France
| | - Sonia André
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1124, Université de Paris, Paris, France
| | - Claire Lozano
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Thierry Vincent
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Laurent Muller
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Jean-Yves Lefrant
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Claire Roger
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Pierre-Géraud Claret
- Medical and Surgical Emergency Department, Nîmes University Hospital, Nîmes, France
| | - Sandra Duvnjak
- Gerontology Department, Nîmes University Hospital, Nîmes, France
| | - Paul Loubet
- Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Albert Sotto
- Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Tu-Ahn Tran
- Pediatrics Department, Nîmes University Hospital, Nîmes, France
| | - Jérôme Estaquier
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1124, Université de Paris, Paris, France
- Laval University Research Center, Quebec City, QC, Canada
| | - Pierre Corbeau
- Immunology Department, Nîmes University Hospital, Nîmes, France
- Institute of Human Genetics, UMR9002, Centre National de la Recherche Scientifique (CNRS) and Montpellier University, Montpellier, France
| |
Collapse
|
34
|
Yazdanpanah N, Sedikides C, Ochs HD, Camargo CA, Darmstadt GL, Cerda A, Cauda V, Peters GJ, Sellke F, Wong ND, Comini E, Jimeno AR, Glover V, Hatziargyriou N, Vincenot CE, Bordas SPA, Rao IM, Abolhassani H, Gharehpetian GB, Weiskirchen R, Gupta M, Chandel SS, Olusanya BO, Cheson B, Pomponio A, Tanzer M, Myles PS, Ma WX, Bella F, Ghavami S, Moein Moghimi S, Pratico D, Hernandez AM, Martinez-Urbistondo M, Urbistondo DM, Fereshtehnejad SM, Ali I, Kimura S, Wallace Hayes A, Cai W, Ernest CKJ, Thomas S, Rahimi K, Sorooshian A, Schreiber M, Kato K, Luong JHT, Pluchino S, Lozano AM, Seymour JF, Kosik KS, Hofmann SG, McIntyre RS, Perc M, Leemans A, Klein RS, Ogino S, Wlezien C, Perry G, Nieto JJ, Levin L, Klionsky DJ, Mobasher B, Dorigo T, Rezaei N. Global Challenges After a Global Challenge: Lessons Learned from the COVID-19 Pandemic. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1457:1-31. [PMID: 39283418 DOI: 10.1007/978-3-031-61939-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Coronavirus disease 2019 (COVID-19) has affected not only individual lives but also the world and global systems, both natural and human-made. Besides millions of deaths and environmental challenges, the rapid spread of the infection and its very high socioeconomic impact have affected healthcare, economic status and wealth, and mental health across the globe. To better appreciate the pandemic's influence, multidisciplinary and interdisciplinary approaches are needed. In this chapter, world-leading scientists from different backgrounds share collectively their views about the pandemic's footprint and discuss challenges that face the international community.
Collapse
Affiliation(s)
- Niloufar Yazdanpanah
- , Houston, USA
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hans D Ochs
- , Houston, USA
- Department of Pediatrics, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, WA, USA
| | - Carlos A Camargo
- , Houston, USA
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Gary L Darmstadt
- , Houston, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Artemi Cerda
- , Houston, USA
- Soil Erosion and Degradation Research Group, Department of Geography, Valencia University, Blasco Ibàñez, Valencia, Spain
| | - Valentina Cauda
- , Houston, USA
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi, Turin, Italy
| | - Godefridus J Peters
- , Houston, USA
- Laboratory Medical Oncology, Amsterdam University Medical Centers, Location VUMC, Amsterdam, the Netherlands
- Department of Biochemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Frank Sellke
- , Houston, USA
- Warren Alpert Medical School, Brown University, Providence, RI, USA
- Division of Cardiothoracic Surgery, Rhode Island Hospital, Providence, RI, USA
| | - Nathan D Wong
- , Houston, USA
- Heart Disease Prevention Program, Division of Cardiology, University of California Irvine, C-240 Medical Sciences, Irvine, CA, USA
| | - Elisabetta Comini
- , Houston, USA
- SENSOR Laboratory, University of Brescia, Brescia, Italy
| | - Alberto Ruiz Jimeno
- , Houston, USA
- Instituto de Física de Cantabria (IFCA), CSIC-Universidad de Cantabria, Santander, Spain
| | - Vivette Glover
- , Houston, USA
- Department of Metabolism, Digestion and Reproduction Hammersmith Hospital Campus, Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| | - Nikos Hatziargyriou
- , Houston, USA
- School of Electrical and Computer Engineering, National Technical University of Athens (NTUA), Athens, Greece
| | - Christian E Vincenot
- , Houston, USA
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Grand Duchy of Luxembourg
| | - Stéphane P A Bordas
- , Houston, USA
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Grand Duchy of Luxembourg
| | - Idupulapati M Rao
- , Houston, USA
- Alliance of Bioversity International, International Center for Tropical Agriculture, Cali, Colombia
- International Centre of Insect Physiology and Ecology (ICIPE), Nairobi, Kenya
| | - Hassan Abolhassani
- , Houston, USA
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | | | - Ralf Weiskirchen
- , Houston, USA
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Manoj Gupta
- , Houston, USA
- Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
| | - Shyam Singh Chandel
- , Houston, USA
- Photovoltaics Research Group, Centre of Excellence in Energy Science and Technology, Shoolini University, Solan, Himachal Pradesh, 173212, India
| | | | - Bruce Cheson
- , Houston, USA
- Center for Cancer and Blood Disorders, Bethesda, MD, USA
| | - Alessio Pomponio
- , Houston, USA
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Bari, Italy
| | - Michael Tanzer
- , Houston, USA
- Division of Orthopedic Surgery, McGill University, Montreal, QC, Canada
| | - Paul S Myles
- , Houston, USA
- Alfred Hospital and Monash University, Melbourne, Australia
| | - Wen-Xiu Ma
- , Houston, USA
- Department of Mathematics and Statistics, University of South Florida, Tampa, FL, USA
- Department of Mathematics, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Material Science Innovation and Modelling, North-West University, Mafikeng Campus, Mmabatho, 2735, South Africa
| | - Federico Bella
- , Houston, USA
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi, Turin, Italy
| | - Saeid Ghavami
- , Houston, USA
- Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - S Moein Moghimi
- , Houston, USA
- School of Pharmacy, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
- Faculty of Health and Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Domenico Pratico
- , Houston, USA
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Alfredo M Hernandez
- , Houston, USA
- Medicine and Endocrinology Department, Universidad de Valladolid and IMDEA, Madrid, Spain
| | | | | | - Seyed-Mohammad Fereshtehnejad
- , Houston, USA
- Division of Neurology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden
| | - Imran Ali
- , Houston, USA
- Department of Chemistry, Jamia Millia Islamia (Central University), New Delhi, India
| | - Shinya Kimura
- , Houston, USA
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - A Wallace Hayes
- , Houston, USA
- Center for Environmental/Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, FL, 33612, USA
- Michigan State University, East Lansing, MI, USA
| | - Wenju Cai
- , Houston, USA
- CSIRO Environment, Hobart, TAS, Australia
| | - Chua K J Ernest
- , Houston, USA
- Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
| | - Sabu Thomas
- , Houston, USA
- School of Energy Materials, Mahatma Gandhi University, Kottayam, Kerala, India
| | - Kazem Rahimi
- , Houston, USA
- Deep Medicine, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Armin Sorooshian
- , Houston, USA
- Department of Chemical and Environmental Engineering, University of Arizona, Tucson, AZ, USA
| | - Michael Schreiber
- , Houston, USA
- Institut für Physik, Technische Universität Chemnitz, 09107, Chemnitz, Germany
| | - Koichi Kato
- , Houston, USA
- Exploratory Research Center on Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - John H T Luong
- , Houston, USA
- School of Chemistry, University College Cork, Cork, T12 YN60, Ireland
| | - Stefano Pluchino
- , Houston, USA
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Andres M Lozano
- , Houston, USA
- Division of Neurosurgery, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, Toronto, ON, Canada
| | - John F Seymour
- , Houston, USA
- Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Kenneth S Kosik
- , Houston, USA
- Department of Molecular Cellular Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - Stefan G Hofmann
- , Houston, USA
- Department of Psychology, Philipps-University Marburg, Marburg, Germany
| | - Roger S McIntyre
- , Houston, USA
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Matjaz Perc
- , Houston, USA
- Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, Maribor, Slovenia
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 404332, Taiwan
- Alma Mater Europaea, Slovenska ulica 17, 2000, Maribor, Slovenia
- Complexity Science Hub Vienna, Josefstädterstraße 39, 1080, Vienna, Austria
- Department of Physics, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea
| | - Alexander Leemans
- , Houston, USA
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Robyn S Klein
- , Houston, USA
- Center for Neuroimmunology and Neuroinfectious Diseases, St. Louis, MO, USA
- Departments of Medicine, Pathology and Immunology, and Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Shuji Ogino
- , Houston, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christopher Wlezien
- , Houston, USA
- Department of Government, University of Texas at Austin, Austin, TX, USA
| | - George Perry
- , Houston, USA
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Juan J Nieto
- , Houston, USA
- CITMAga, University of Santiago de Compostela, A Coruña, Spain
| | - Lisa Levin
- , Houston, USA
- Center for Marine Biodiversity and Conservation, Integrative Oceanography Division, Scripps Institution of Oceanography, University of California, San Diego, San Diego, CA, USA
| | - Daniel J Klionsky
- , Houston, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Bahram Mobasher
- , Houston, USA
- Department of Physics and Astronomy, University of California, Riverside, CA, USA
| | - Tommaso Dorigo
- , Houston, USA
- Lulea University of Technology, Laboratorievagen 14, Lulea, Sweden
- Istituto Nazionale di Fisica Nucleare (INFN), Via Francesco Marzolo, Sezione di Padova, Italy
| | - Nima Rezaei
- , Houston, USA.
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
35
|
Harte JV, Coleman-Vaughan C, Crowley MP, Mykytiv V. It's in the blood: a review of the hematological system in SARS-CoV-2-associated COVID-19. Crit Rev Clin Lab Sci 2023; 60:595-624. [PMID: 37439130 DOI: 10.1080/10408363.2023.2232010] [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: 04/10/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to an unprecedented global healthcare crisis. While SARS-CoV-2-associated COVID-19 affects primarily the respiratory system, patients with COVID-19 frequently develop extrapulmonary manifestations. Notably, changes in the hematological system, including lymphocytopenia, neutrophilia and significant abnormalities of hemostatic markers, were observed early in the pandemic. Hematological manifestations have since been recognized as important parameters in the pathophysiology of SARS-CoV-2 and in the management of patients with COVID-19. In this narrative review, we summarize the state-of-the-art regarding the hematological and hemostatic abnormalities observed in patients with SARS-CoV-2-associated COVID-19, as well as the current understanding of the hematological system in the pathophysiology of acute and chronic SARS-CoV-2-associated COVID-19.
Collapse
Affiliation(s)
- James V Harte
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
- School of Biochemistry & Cell Biology, University College Cork, Cork, Ireland
| | | | - Maeve P Crowley
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
- Irish Network for Venous Thromboembolism Research (INViTE), Ireland
| | - Vitaliy Mykytiv
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
| |
Collapse
|
36
|
Riegler AN, Benson P, Long K, Leal SM. Differential activation of programmed cell death in patients with severe SARS-CoV-2 infection. Cell Death Discov 2023; 9:420. [PMID: 37985756 PMCID: PMC10662024 DOI: 10.1038/s41420-023-01715-4] [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: 09/05/2023] [Revised: 10/26/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes severe lower airway disease and death in a subset of patients. Knowledge on the relative contribution of programmed cell death (PCD) to lung pathology is limited to few human autopsy studies with small sample size/scope, in vitro cell culture, and experimental model systems. In this study, we sought to identify, localize, and quantify activation of apoptosis, ferroptosis, pyroptosis, and necroptosis in FFPE lung tissues from patients that died from severe SARS-CoV-2 infection (n = 28) relative to uninfected controls (n = 13). Immunofluorescence (IF) staining, whole-slide imaging, and Image J software was used to localize and quantify expression of SARS-CoV-2 nucleoprotein and the following PCD protein markers: cleaved Caspase-3, pMLKL, cleaved Gasdermin D, and CD71, respectively. IF showed differential activation of each PCD pathway in infected lungs and dichotomous staining for SARS-CoV-2 nucleoprotein enabling distinction between high (n = 9) vs low viral burden (n = 19). No differences were observed in apoptosis and ferroptosis in SARS-CoV-2 infected lungs relative to uninfected controls. However, both pyroptosis and necroptosis were significantly increased in SARS-CoV-2-infected lungs. Increased pyroptosis was observed in SARS-CoV-2 infected lungs, irrespective of viral burden, suggesting an inflammation-driven mechanism. In contrast, necroptosis exhibited a very strong positive correlation with viral burden (R2 = 0.9925), suggesting a direct SARS-CoV-2 mediated effect. These data indicate a possible novel mechanism for viral-mediated necroptosis and a potential role for both lytic programmed cell death pathways, necroptosis and pyroptosis, in mediating infection outcome.
Collapse
Affiliation(s)
- Ashleigh N Riegler
- Division of Laboratory Medicine, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Paul Benson
- Division of Anatomic Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth Long
- Division of Infectious Diseases, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sixto M Leal
- Division of Laboratory Medicine, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
37
|
Wang B, Chen Z, Huang Y, Ding J, Lin Y, Wang M, Li X. Mitochondrial mass of circulating NK cells as a novel biomarker in severe SARS-CoV-2 infection. Int Immunopharmacol 2023; 124:110839. [PMID: 37639852 DOI: 10.1016/j.intimp.2023.110839] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/05/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Severe SARS-CoV-2 infection results in lymphopenia and impaired function of T, B, and NK (TBNK-dominant) lymphocytes. Mitochondria are essential targets of SARS-CoV-2 and the efficacy of lymphocyte mitochondrial function for immunosurveillance in COVID-19 patients has not been evaluated. METHODS Multi-parametric flow cytometry was used to characterize mitochondrial function, including mitochondrial mass (MM) and low mitochondrial membrane potential (MMPlow), in TBNK-dominant lymphocytes from severe (n = 93) and moderate (n = 77) hospitalized COVID-19 patients. We compared the role of novel lymphocyte mitochondrial indicators and routine infection biomarkers as early predictors of severity and death in COVID-19 patients. We then developed a mortality decision tree prediction model based on immunosurveillance indicators through machine learning. RESULTS At admission, the MM of circulating NK cells (NK-MM) was the best discriminator of severe/moderate disease (AUC = 0.8067) compared with the routine infection biomarkers. The NK cell count and NK-MM displayed superior diagnostic effects to distinguish patients with non-fatal or fatal outcomes. Interestingly, NK-MM was significantly polarized in non-survivors, with some patients showing a decrease and others showing an abnormal increase. Kaplan-Meier analysis showed that NK-MM had the optimal predictive efficacy (hazard ratio = 11.66). The decision tree model has the highest proportion of importance for NK-MM, which is superior to the single diagnostic effect of the above indicators (AUC = 0.8900). CONCLUSION NK-MM was not only associated with disease severity, its abnormal increases or decreases also predicted mortality risk. The resulting decision tree prediction model is the first to focus on immune monitoring indicators to provide decision-making clues for COVID-19 clinical management.
Collapse
Affiliation(s)
- Bingqi Wang
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhenni Chen
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yiran Huang
- School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jiayi Ding
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yingrui Lin
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Min Wang
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xianping Li
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
38
|
Gałgańska H, Jarmuszkiewicz W, Gałgański Ł. Carbon dioxide and MAPK signalling: towards therapy for inflammation. Cell Commun Signal 2023; 21:280. [PMID: 37817178 PMCID: PMC10566067 DOI: 10.1186/s12964-023-01306-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/05/2023] [Indexed: 10/12/2023] Open
Abstract
Inflammation, although necessary to fight infections, becomes a threat when it exceeds the capability of the immune system to control it. In addition, inflammation is a cause and/or symptom of many different disorders, including metabolic, neurodegenerative, autoimmune and cardiovascular diseases. Comorbidities and advanced age are typical predictors of more severe cases of seasonal viral infection, with COVID-19 a clear example. The primary importance of mitogen-activated protein kinases (MAPKs) in the course of COVID-19 is evident in the mechanisms by which cells are infected with SARS-CoV-2; the cytokine storm that profoundly worsens a patient's condition; the pathogenesis of diseases, such as diabetes, obesity, and hypertension, that contribute to a worsened prognosis; and post-COVID-19 complications, such as brain fog and thrombosis. An increasing number of reports have revealed that MAPKs are regulated by carbon dioxide (CO2); hence, we reviewed the literature to identify associations between CO2 and MAPKs and possible therapeutic benefits resulting from the elevation of CO2 levels. CO2 regulates key processes leading to and resulting from inflammation, and the therapeutic effects of CO2 (or bicarbonate, HCO3-) have been documented in all of the abovementioned comorbidities and complications of COVID-19 in which MAPKs play roles. The overlapping MAPK and CO2 signalling pathways in the contexts of allergy, apoptosis and cell survival, pulmonary oedema (alveolar fluid resorption), and mechanical ventilation-induced responses in lungs and related to mitochondria are also discussed. Video Abstract.
Collapse
Affiliation(s)
- Hanna Gałgańska
- Faculty of Biology, Molecular Biology Techniques Laboratory, Adam Mickiewicz University in Poznan, Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland
| | - Wieslawa Jarmuszkiewicz
- Faculty of Biology, Department of Bioenergetics, Adam Mickiewicz University in Poznan, Institute of Molecular Biology and Biotechnology, Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland
| | - Łukasz Gałgański
- Faculty of Biology, Department of Bioenergetics, Adam Mickiewicz University in Poznan, Institute of Molecular Biology and Biotechnology, Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland.
| |
Collapse
|
39
|
Shoraka S, Mohebbi SR, Hosseini SM, Zali MR. Comparison of plasma mitochondrial DNA copy number in asymptomatic and symptomatic COVID-19 patients. Front Microbiol 2023; 14:1256042. [PMID: 37869674 PMCID: PMC10587688 DOI: 10.3389/fmicb.2023.1256042] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction Since the beginning of the COVID-19 pandemic, a wide clinical spectrum, from asymptomatic infection to mild or severe disease and death, have been reported in COVID-19 patients. Studies have suggested several possible factors, which may affect the clinical outcome of COVID-19. A pro-inflammatory state and impaired antiviral response have been suggested as major contributing factors in severe COVID-19. Considering that mitochondria have an important role in regulating the immune responses to pathogens, pro-inflammatory signaling, and cell death, it has received much attention in SARS-CoV-2 infection. Recent studies have demonstrated that high levels of cell-free mitochondrial DNA (cf-mtDNA) are associated with an increased risk of COVID-19 intensive care unit (ICU) admission and mortality. However, there have been few studies on cf-mtDNA in SARS-CoV-2 infection, mainly focusing on critically ill COVID-19 cases. In the present study, we investigated cf-mtDNA copy number in COVID-19 patients and compared between asymptomatic and symptomatic cases, and assessed the clinical values. We also determined the cf-nuclear DNA (cf-nDNA) copy number and mitochondrial transcription factor A (TFAM) mRNA level in the studied groups. Materials and methods Plasma and buffy coat samples were collected from 37 COVID-19 patients and 33 controls. Briefly, after total DNA extraction, plasma cf-mtDNA, and cf-nDNA copy numbers were measured by absolute qPCR using a standard curve method. Furthermore, after total RNA extraction from buffy coat and cDNA synthesis, TFAM mRNA levels were evaluated by qPCR. Results The results showed that cf-mtDNA levels in asymptomatic COVID-19 patients were statistically significantly higher than in symptomatic cases (p value = 0.01). However, cf-nDNA levels were higher in symptomatic patients than in asymptomatic cases (p value = 0.00). There was no significant difference between TFAM levels in the buffy coat of these two groups (p value > 0.05). Also, cf-mtDNA levels showed good diagnostic potential in COVID-19 subgroups. Conclusion cf-mtDNA is probably important in the outcome of SARS-CoV-2 infection due to its role in inflammation and immune response. It can also be a promising candidate biomarker for the diagnosis of COVID-19 subgroups. Further investigation will help understanding the COVID-19 pathophysiology and effective diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Shahrzad Shoraka
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Seyed Reza Mohebbi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Masoud Hosseini
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Jain R, Mathew D. Mechanisms influencing the high prevalence of COVID-19 in diabetics: A systematic review. MEDICAL RESEARCH ARCHIVES 2023; 11:4540. [PMID: 38933091 PMCID: PMC11198970 DOI: 10.18103/mra.v11i10.4540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Diabetics have an increased risk of contracting COVID-19 infection and tend to have more severe symptoms. This systematic review explores the potential mechanisms influencing the high prevalence of COVID-19 infections in individuals with diabetes. It reviews the emerging evidence about the interactions between viral and diabetic pathways, particularly how diabetes physiology could contribute to higher viral reception, viral entry and pathogenicity, and the severity of disease symptoms. Finally, it examines the challenges we face in studying these mechanisms and offers new strategies that might assist our fight against current and future pandemics.
Collapse
Affiliation(s)
- Roshni Jain
- Cell and Molecular Biology Program, University of Nevada, Reno, NV 89557
- Department of Biology, University of Nevada, Reno, NV 89557
| | - Dennis Mathew
- Cell and Molecular Biology Program, University of Nevada, Reno, NV 89557
- Department of Biology, University of Nevada, Reno, NV 89557
| |
Collapse
|
41
|
Zhang D, Jia M, Wang C, Li Y, Ma C, Zhu G, Ma R, Wen D, Jia X, Xu G, Zhang X, Cong B. CCK2-receptor deficiency impairs immune balance by influencing CD4 + T cells development by inhibiting cortical-thymic-epithelial-cells. Exp Biol Med (Maywood) 2023; 248:1718-1731. [PMID: 37787155 PMCID: PMC10792431 DOI: 10.1177/15353702231198083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/05/2023] [Indexed: 10/04/2023] Open
Abstract
Immune balance is crucial for an organism's survival and is inseparable from the regulation of the nervous system. Accumulating evidence indicates that cholecystokinin (CCK) plays an important role in mediating the immune response through the activation of cholecystokinin receptors (CCKRs). However, it remains unclear whether CCKRs deficiency may impair immune balance. Here, we showed that CCK2R-deficient adult mice were immunocompromised and had an increased risk of shock and even death in an endotoxemia (ETM)/endotoxin shock (ES) model. In addition, in both adult and juvenile mice, CCK2R deficiency not only influenced the development of CD4 single-positive (SP) thymocytes in thymic positive selection but also decreased the population of CD3+ CD4+ T cells in the spleen. More importantly, CCK2R deficiency inhibited the expression of major histocompatibility complex class II (MHC II) and CD83 on cortical thymic epithelial cells (cTECs) in juvenile and adult mice. Overall, our study suggests that CCK2R is essential for maintaining CD4+ T cell development in the thymus and reveals that CCK2R plays an important role in maintaining immune balance.
Collapse
Affiliation(s)
- Dong Zhang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China
- Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050017, China
| | - Miaomiao Jia
- Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China
| | - Chuan Wang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China
| | - Yingmin Li
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China
| | - Guiyun Zhu
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China
- Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China
| | - Rufei Ma
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China
| | - Di Wen
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China
| | - Xianxian Jia
- Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China
| | - Guangming Xu
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaojing Zhang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China
| | - Bin Cong
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China
- Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China
| |
Collapse
|
42
|
Yuan C, Ma Z, Xie J, Li W, Su L, Zhang G, Xu J, Wu Y, Zhang M, Liu W. The role of cell death in SARS-CoV-2 infection. Signal Transduct Target Ther 2023; 8:357. [PMID: 37726282 PMCID: PMC10509267 DOI: 10.1038/s41392-023-01580-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/09/2023] [Accepted: 07/31/2023] [Indexed: 09/21/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), showing high infectiousness, resulted in an ongoing pandemic termed coronavirus disease 2019 (COVID-19). COVID-19 cases often experience acute respiratory distress syndrome, which has caused millions of deaths. Apart from triggering inflammatory and immune responses, many viral infections can cause programmed cell death in infected cells. Cell death mechanisms have a vital role in maintaining a suitable environment to achieve normal cell functionality. Nonetheless, these processes are dysregulated, potentially contributing to disease pathogenesis. Over the past decades, multiple cell death pathways are becoming better understood. Growing evidence suggests that the induction of cell death by the coronavirus may significantly contributes to viral infection and pathogenicity. However, the interaction of SARS-CoV-2 with cell death, together with its associated mechanisms, is yet to be elucidated. In this review, we summarize the existing evidence concerning the molecular modulation of cell death in SARS-CoV-2 infection as well as viral-host interactions, which may shed new light on antiviral therapy against SARS-CoV-2.
Collapse
Affiliation(s)
- Cui Yuan
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Zhenling Ma
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Jiufeng Xie
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Wenqing Li
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Lijuan Su
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Guozhi Zhang
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Jun Xu
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Yaru Wu
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Min Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Wei Liu
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China.
| |
Collapse
|
43
|
Yu H, Yuan L, Yan Z, Zhou M, Ye J, Wu K, Chen W, Chen R, Xia N, Guan Y, Zhu H. Butyrate Protects against SARS-CoV-2-Induced Tissue Damage in Golden Hamsters. Int J Mol Sci 2023; 24:14191. [PMID: 37762492 PMCID: PMC10532055 DOI: 10.3390/ijms241814191] [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: 08/11/2023] [Revised: 09/09/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Butyrate, produced by gut microbe during dietary fiber fermentation, has anti-inflammatory and antioxidant effects on chronic inflammation diseases, yet it remains to be explored whether butyrate has protective effects against viral infections. Here, we demonstrated that butyrate alleviated tissue injury in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected golden hamsters supplemented with butyrate before and during the infection. Butyrate-treated hamsters showed augmentation of type I interferon (IFN) response and activation of endothelial cells without exaggerated inflammation. In addition, butyrate regulated redox homeostasis by enhancing the activity of superoxide dismutase (SOD) to inhibit excessive apoptotic cell death. Therefore, butyrate exhibited effective prevention against SARS-CoV-2 by upregulating antiviral immune responses and promoting cell survival.
Collapse
Affiliation(s)
- Huan Yu
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
| | - Lunzhi Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhigang Yan
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
| | - Ming Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jianghui Ye
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Kun Wu
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wenjia Chen
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
| | - Rirong Chen
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yi Guan
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
- State Key Laboratory of Emerging Infectious Diseases (SKLEID), School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- EKIH (Gewuzhikang) Advanced Pathogen Research Institute, Futian District, Shenzhen 518045, China
| | - Huachen Zhu
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
- State Key Laboratory of Emerging Infectious Diseases (SKLEID), School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- EKIH (Gewuzhikang) Advanced Pathogen Research Institute, Futian District, Shenzhen 518045, China
| |
Collapse
|
44
|
Hovhannisyan G, Harutyunyan T, Aroutiounian R, Liehr T. The Diagnostic, Prognostic, and Therapeutic Potential of Cell-Free DNA with a Special Focus on COVID-19 and Other Viral Infections. Int J Mol Sci 2023; 24:14163. [PMID: 37762464 PMCID: PMC10532175 DOI: 10.3390/ijms241814163] [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: 08/02/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Cell-free DNA (cfDNA) in human blood serum, urine, and other body fluids recently became a commonly used diagnostic marker associated with various pathologies. This is because cfDNA enables a much higher sensitivity than standard biochemical parameters. The presence of and/or increased level of cfDNA has been reported for various diseases, including viral infections, including COVID-19. Here, we review cfDNA in general, how it has been identified, where it can derive from, its molecular features, and mechanisms of release and clearance. General suitability of cfDNA for diagnostic questions, possible shortcomings and future directions are discussed, with a special focus on coronavirus infection.
Collapse
Affiliation(s)
- Galina Hovhannisyan
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (G.H.); (T.H.); (R.A.)
| | - Tigran Harutyunyan
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (G.H.); (T.H.); (R.A.)
| | - Rouben Aroutiounian
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (G.H.); (T.H.); (R.A.)
| | - Thomas Liehr
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
45
|
Peleman C, Van Coillie S, Ligthart S, Choi SM, De Waele J, Depuydt P, Benoit D, Schaubroeck H, Francque SM, Dams K, Jacobs R, Robert D, Roelandt R, Seurinck R, Saeys Y, Rajapurkar M, Jorens PG, Hoste E, Vanden Berghe T. Ferroptosis and pyroptosis signatures in critical COVID-19 patients. Cell Death Differ 2023; 30:2066-2077. [PMID: 37582864 PMCID: PMC10482958 DOI: 10.1038/s41418-023-01204-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/26/2023] [Accepted: 08/04/2023] [Indexed: 08/17/2023] Open
Abstract
Critical COVID-19 patients admitted to the intensive care unit (ICU) frequently suffer from severe multiple organ dysfunction with underlying widespread cell death. Ferroptosis and pyroptosis are two detrimental forms of regulated cell death that could constitute new therapeutic targets. We enrolled 120 critical COVID-19 patients in a two-center prospective cohort study to monitor systemic markers of ferroptosis, iron dyshomeostasis, pyroptosis, pneumocyte cell death and cell damage on the first three consecutive days after ICU admission. Plasma of 20 post-operative ICU patients (PO) and 39 healthy controls (HC) without organ failure served as controls. Subsets of COVID-19 patients displayed increases in individual biomarkers compared to controls. Unsupervised clustering was used to discern latent clusters of COVID-19 patients based on biomarker profiles. Pyroptosis-related interleukin-18 accompanied by high pneumocyte cell death was independently associated with higher odds at mechanical ventilation, while the subgroup with high interleuking-1 beta (but limited pneumocyte cell death) displayed reduced odds at mechanical ventilation and lower mortality hazard. Meanwhile, iron dyshomeostasis with a tendency towards higher ferroptosis marker malondialdehyde had no association with outcome, except for the small subset of patients with very high catalytic iron independently associated with reduced survival. Forty percent of patients did not have a clear signature of the cell death mechanisms studied in this cohort. Moreover, repeated moderate levels of soluble receptor of advanced glycation end products and growth differentiation factor 15 during the first three days after ICU admission are independently associated with adverse clinical outcome compared to sustained lower levels. Altogether, the data point towards distinct subgroups in this cohort of critical COVID-19 patients with different systemic signatures of pyroptosis, iron dyshomeostasis, ferroptosis or pneumocyte cell death markers that have different outcomes in ICU. The distinct groups may allow 'personalized' treatment allocation in critical COVID-19 based on systemic biomarker profiles.
Collapse
Affiliation(s)
- Cédric Peleman
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Samya Van Coillie
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Symen Ligthart
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Intensive Care, Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Sze Men Choi
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jan De Waele
- Intensive Care Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Pieter Depuydt
- Intensive Care Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Dominique Benoit
- Intensive Care Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Hannah Schaubroeck
- Intensive Care Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Sven M Francque
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Karolien Dams
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Intensive Care, Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Rita Jacobs
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Intensive Care, Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Dominique Robert
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Intensive Care, Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Ria Roelandt
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Ruth Seurinck
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Yvan Saeys
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Mohan Rajapurkar
- Department of Nephrology, Muljibhai Patel Society for Research in Nephro-Urology, Nadiad, India
| | - Philippe G Jorens
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Intensive Care, Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Eric Hoste
- Intensive Care Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
46
|
Aiello A, Najafi-Fard S, Goletti D. Initial immune response after exposure to Mycobacterium tuberculosis or to SARS-COV-2: similarities and differences. Front Immunol 2023; 14:1244556. [PMID: 37662901 PMCID: PMC10470049 DOI: 10.3389/fimmu.2023.1244556] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) and Coronavirus disease-2019 (COVID-19), whose etiologic agent is severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), are currently the two deadliest infectious diseases in humans, which together have caused about more than 11 million deaths worldwide in the past 3 years. TB and COVID-19 share several aspects including the droplet- and aerosol-borne transmissibility, the lungs as primary target, some symptoms, and diagnostic tools. However, these two infectious diseases differ in other aspects as their incubation period, immune cells involved, persistence and the immunopathological response. In this review, we highlight the similarities and differences between TB and COVID-19 focusing on the innate and adaptive immune response induced after the exposure to Mtb and SARS-CoV-2 and the pathological pathways linking the two infections. Moreover, we provide a brief overview of the immune response in case of TB-COVID-19 co-infection highlighting the similarities and differences of each individual infection. A comprehensive understanding of the immune response involved in TB and COVID-19 is of utmost importance for the design of effective therapeutic strategies and vaccines for both diseases.
Collapse
Affiliation(s)
| | | | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
47
|
Wang Y, Yang C, Wang Z, Wang Y, Yan Q, Feng Y, Liu Y, Huang J, Zhou J. Epithelial Galectin-3 Induced the Mitochondrial Complex Inhibition and Cell Cycle Arrest of CD8 + T Cells in Severe/Critical COVID-19. Int J Mol Sci 2023; 24:12780. [PMID: 37628961 PMCID: PMC10454470 DOI: 10.3390/ijms241612780] [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: 06/29/2023] [Revised: 07/20/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Previous research suggested that the dramatical decrease in CD8+ T cells is a contributing factor in the poor prognosis and disease progression of COVID-19 patients. However, the underlying mechanisms are not fully understood. In this study, we conducted Single-cell RNA sequencing (scRNA-seq) and single-cell T cell receptor sequencing (scTCR-seq) analysis, which revealed a proliferative-exhausted MCM+FASLGlow CD8+ T cell phenotype in severe/critical COVID-19 patients. These CD8+ T cells were characterized by G2/M cell cycle arrest, downregulation of respiratory chain complex genes, and inhibition of mitochondrial biogenesis. CellChat analysis of infected lung epithelial cells and CD8+ T cells found that the galectin signaling pathway played a crucial role in CD8+ T cell reduction and dysfunction. To further elucidate the mechanisms, we established SARS-CoV-2 ORF3a-transfected A549 cells, and co-cultured them with CD8+ T cells for ex vivo experiments. Our results showed that epithelial galectin-3 inhibited the transcription of the mitochondrial respiratory chain complex III/IV genes of CD8+ T cells by suppressing the nuclear translocation of nuclear respiratory factor 1 (NRF1). Further findings showed that the suppression of NRF1 translocation was associated with ERK-related and Akt-related signaling pathways. Importantly, the galectin-3 inhibitor, TD-139, promoted nuclear translocation of NRF1, thus enhancing the expression of the mitochondrial respiratory chain complex III/IV genes and the mitochondrial biogenesis of CD8+ T cells. Our study provided new insights into the immunopathogenesis of COVID-19 and identified potential therapeutic targets for the prevention and treatment of severe/critical COVID-19 patients.
Collapse
Affiliation(s)
- Yudie Wang
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Cheng Yang
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhongyi Wang
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yi Wang
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qing Yan
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ying Feng
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yanping Liu
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Juan Huang
- Department of Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China
| | - Jingjiao Zhou
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| |
Collapse
|
48
|
Azarias Da Silva M, Nioche P, Soudaramourty C, Bull-Maurer A, Tiouajni M, Kong D, Zghidi-Abouzid O, Picard M, Mendes-Frias A, Santa-Cruz A, Carvalho A, Capela C, Pedrosa J, Castro AG, Loubet P, Sotto A, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Tran TA, Tokunaga K, Silvestre R, Corbeau P, Mammano F, Estaquier J. Repetitive mRNA vaccination is required to improve the quality of broad-spectrum anti-SARS-CoV-2 antibodies in the absence of CXCL13. SCIENCE ADVANCES 2023; 9:eadg2122. [PMID: 37540749 PMCID: PMC10403221 DOI: 10.1126/sciadv.adg2122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/05/2023] [Indexed: 08/06/2023]
Abstract
Since the initial spread of severe acute respiratory syndrome coronavirus 2 infection, several viral variants have emerged and represent a major challenge for immune control, particularly in the context of vaccination. We evaluated the quantity, quality, and persistence of immunoglobulin G (IgG) and IgA in individuals who received two or three doses of messenger RNA (mRNA) vaccines, compared with previously infected vaccinated individuals. We show that three doses of mRNA vaccine were required to match the humoral responses of preinfected vaccinees. Given the importance of antibody-dependent cell-mediated immunity against viral infections, we also measured the capacity of IgG to recognize spike variants expressed on the cell surface and found that cross-reactivity was also strongly improved by repeated vaccination. Last, we report low levels of CXCL13, a surrogate marker of germinal center activation and formation, in vaccinees both after two and three doses compared with preinfected individuals, providing a potential explanation for the short duration and low quality of Ig induced.
Collapse
Affiliation(s)
| | - Pierre Nioche
- INSERM-U1124, Université Paris Cité, Paris, France
- Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | | | | | - Mounira Tiouajni
- INSERM-U1124, Université Paris Cité, Paris, France
- Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | - Dechuan Kong
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | - Ana Mendes-Frias
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - André Santa-Cruz
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal
| | - Alexandre Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal
| | - Carlos Capela
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal
| | - Jorge Pedrosa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António Gil Castro
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Paul Loubet
- Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Albert Sotto
- Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Laurent Muller
- Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | | | - Claire Roger
- Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | | | - Sandra Duvnjak
- Service de Gérontologie et Prévention du Vieillissement, CHU de Nîmes, Nîmes, France
| | - Tu-Anh Tran
- Service de Pédiatrie, CHU de Nîmes, Nîmes, France
| | - Kenzo Tokunaga
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Pierre Corbeau
- Institut de Génétique Humaine, UMR9002 CNRS-Université de Montpellier, Montpellier, France
- Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France
| | - Fabrizio Mammano
- INSERM-U1124, Université Paris Cité, Paris, France
- Université de Tours, INSERM, UMR1259 MAVIVH, Tours, France
| | - Jérôme Estaquier
- INSERM-U1124, Université Paris Cité, Paris, France
- CHU de Québec-Université Laval Research Center, Québec City, Québec, Canada
| |
Collapse
|
49
|
Heng WT, Lim HX, Tan KO, Poh CL. Validation of Multi-epitope Peptides Encapsulated in PLGA Nanoparticles Against Influenza A Virus. Pharm Res 2023; 40:1999-2025. [PMID: 37344603 DOI: 10.1007/s11095-023-03540-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/19/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND Influenza is a highly contagious respiratory disease which poses a serious threat to public health globally, causing severe diseases in 3-5 million humans and resulting in 650,000 deaths annually. The current licensed seasonal influenza vaccines lacked cross-reactivity against novel emerging influenza strains as they conferred limited neutralising capabilities. To address the issue, we designed a multi-epitope peptide-based vaccine delivered by the self-adjuvanting PLGA nanoparticles against influenza infections. METHODS A total of six conserved peptides representing B- and T-cell epitopes of Influenza A were identified and they were formulated in either incomplete Freund's adjuvant containing CpG ODN 1826 or being encapsulated in PLGA nanoparticles for the evaluation of immunogenicity in BALB/c mice. RESULTS The self-adjuvanting PLGA nanoparticles encapsulating the six conserved peptides were capable of eliciting the highest levels of IgG and IFN- γ producing cells. In addition, the immunogenicity of the six peptides encapsulated in PLGA nanoparticles showed greater humoral and cellular mediated immune responses elicited by the mixture of six naked peptides formulated in incomplete Freund's adjuvant containing CpG ODN 1826 in the immunized mice. Peptide 3 from the mixture of six peptides was found to exert necrotic effect on CD3+ T-cells and this finding indicated that peptide 3 should be removed from the nanovaccine formulation. CONCLUSION The study demonstrated the self-adjuvanting properties of the PLGA nanoparticles as a delivery system without the need for incorporation of toxic and costly conventional adjuvants in multi-epitope peptide-based vaccines.
Collapse
Affiliation(s)
- Wen Tzuen Heng
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Hui Xuan Lim
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Kuan Onn Tan
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
50
|
Lee YJ, Seok SH, Lee NY, Choi HJ, Lee YW, Chang HJ, Hwang JY, On DI, Noh HA, Lee SB, Kwon HK, Yun JW, Shin JS, Seo JY, Nam KT, Lee H, Lee HY, Park JW, Seong JK. Murine Coronavirus Disease 2019 Lethality Is Characterized by Lymphoid Depletion Associated with Suppressed Antigen-Presenting Cell Functionality. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:866-882. [PMID: 37024046 PMCID: PMC10073095 DOI: 10.1016/j.ajpath.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/28/2023] [Accepted: 03/16/2023] [Indexed: 04/08/2023]
Abstract
The disease severity of coronavirus disease 2019 (COVID-19) varies considerably from asymptomatic to serious, with fatal complications associated with dysregulation of innate and adaptive immunity. Lymphoid depletion in lymphoid tissues and lymphocytopenia have both been associated with poor disease outcomes in patients with COVID-19, but the mechanisms involved remain elusive. In this study, human angiotensin-converting enzyme 2 (hACE2) transgenic mouse models susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection were used to investigate the characteristics and determinants of lethality associated with the lymphoid depletion observed in SARS-CoV-2 infection. The lethality of Wuhan SARS-CoV-2 infection in K18-hACE2 mice was characterized by severe lymphoid depletion and apoptosis in lymphoid tissues related to fatal neuroinvasion. The lymphoid depletion was associated with a decreased number of antigen-presenting cells (APCs) and their suppressed functionality below basal levels. Lymphoid depletion with reduced APC function was a specific feature observed in SARS-CoV-2 infection but not in influenza A infection and had the greatest prognostic value for disease severity in murine COVID-19. Comparison of transgenic mouse models resistant and susceptible to SARS-CoV-2 infection revealed that suppressed APC function could be determined by the hACE2 expression pattern and interferon-related signaling. Thus, we demonstrated that lymphoid depletion associated with suppressed APC function characterizes the lethality of COVID-19 mouse models. Our data also suggest a potential therapeutic approach to prevent the severe progression of COVID-19 by enhancing APC functionality.
Collapse
Affiliation(s)
- Yu Jin Lee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, Republic of Korea
| | - Sang Hyeok Seok
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, Republic of Korea
| | - Na Yun Lee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, Republic of Korea
| | - Hee Jin Choi
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, Republic of Korea
| | - Yoon Woo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Hee Jung Chang
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Ji-Yeon Hwang
- Preclinical Research Center, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Da In On
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, Republic of Korea
| | - Hyun Ah Noh
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, Republic of Korea
| | - Su-Bin Lee
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jun-Won Yun
- College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jeon-Soo Shin
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Biomedical Science Institute, and the Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jun-Young Seo
- Severance Biomedical Science Institute, and the Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, and the Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Ho Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, Republic of Korea.
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, Republic of Korea; Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK 21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|