1
|
Duerr GD, Hamiko M, Beer J, Nattermann J, Schafhaus M, Held SAE, Schewe JC, Wittmann M, Kurts C, Zimmer S, Velten M, Heine A. The interplay between COVID-19 and heart disease: Unravelling a complex connection. Life Sci 2025; 370:123524. [PMID: 40044033 DOI: 10.1016/j.lfs.2025.123524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/23/2025] [Accepted: 03/01/2025] [Indexed: 03/20/2025]
Abstract
The intersection of coronavirus (COVID-19) and heart disease has emerged as a critical nexus in the landscape of global health. Individuals with heart disease face elevated risks when infected with Severe Acute Respiratory-Syndrome Coronavirus-type-2 (SARS-CoV-2) leading to COVID-19. The virus can directly affect the heart, resulting in myocarditis, arrhythmias, and heart failure, even in individuals without prior medical cardiac history. Therefore, tools identifying patients with cardiac infestation and predicting disease severity are of utmost importance. This study's unbiased stratification of clinical and immunological parameters of 134 SARS-CoV-2 positive patients revealed clusters of course-severity within the established WHO ordinal severity-scale leading to its summary (SWOSS) into three categories, A-C. PE and SWOSS-C were significantly associated with reduced survival of COVID-19 patients. The previously introduced CD8/Treg/monocyte-ratio which hints at a dysfunctional antiviral immunity associated with poor prognosis could be verified in this larger study population. However, the number of circulating CD14 + HLA-DR+ monocytes represented the most significant predictor for myocardial damage indicated by PE. We used all available data for an unbiased examination of associations and predictions by machine learning algorithms: Predictive markers for PE can be obtained in clinic and may serve as prognostic features. Among numerous parameters, C-reactive protein (CRP) was the most important in determining the presence of PE and SWOSS-category. Prediction of survival was most relevantly influenced by SWOSS-category underlining the benefit of this condensed classification for clinical practice. All AI-revealed prognostic features serve as promising starting-point to gain further understanding of the interplay between COVID-19 and heart disease.
Collapse
Affiliation(s)
- G D Duerr
- Department of Cardiovascular Surgery, University Medical Center, Johannes-Gutenberg University, Mainz, Germany.
| | - M Hamiko
- Department of Cardiac Surgery, University Hospital Bonn, Bonn, Germany
| | - J Beer
- Department of Cardiovascular Surgery, University Medical Center, Johannes-Gutenberg University, Mainz, Germany
| | - J Nattermann
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - M Schafhaus
- Department of Cardiac Surgery, University Hospital Bonn, Bonn, Germany
| | - S A E Held
- Department of Internal Medicine III for Hematology, Oncology, Rheumatology and Immune-Oncology, University Hospital Bonn, Bonn, Germany
| | - J C Schewe
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Medical Center Rostock, Rostock, Germany
| | - M Wittmann
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - C Kurts
- Institute for Experimental Immunology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - S Zimmer
- Department of Internal Medicine II - Cardiology, University Hospital Bonn, Bonn, Germany
| | - M Velten
- Department of Anesthesiology and Pain Management, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - A Heine
- Department of Internal Medicine III for Hematology, Oncology, Rheumatology and Immune-Oncology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
2
|
Bain V, Correa-Silva S, Matsuo OM, Silva-Avelar I, Zheng Y, Rangel-Santos A, Souza Gonçalves G, de Toledo Fink T, Suguita P, Caires O A Ferreira J, Fernandes Ferreira AE, Litvinov N, Andrade Macaferri da Fonseca F, Astley C, Martins F, Carneiro Sampaio M, de Sousa Marques HH, da Silva CAA, Palmeira P, Bádue Pereira MF. Comprehensive Characterization of Innate and Adaptive Immune Profiles in the Pediatric COVID-19 Convalescent Phase. Pediatr Infect Dis J 2025; 44:557-563. [PMID: 40067781 DOI: 10.1097/inf.0000000000004713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
BACKGROUND Knowledge of the effect of SARS-CoV-2 on the innate and adaptive immune responses of children is currently lacking. We investigated the immune profile of recovered pediatric patients 3 to 11 weeks after acute COVID-19. METHODS Children who were previously healthy or had a preexisting chronic disease and had a positive reverse transcription polymerase chain reaction/serology were enrolled (n=23). The control group was composed of 25 patients without COVID-19 paired by age, sex and baseline chronic conditions. We performed immunophenotyping, hematologic and inflammatory markers analysis, cytokines and T-cell receptor excision circle (TREC) quantifications. RESULTS Most COVID-19 convalescent pediatric patients (COVID-19 CPP) had chronic conditions (73.9%), as well as 80% of the controls. Five children developed multisystem inflammatory syndrome in children. COVID-19 CPP had higher lymphocyte numbers than controls due to an increase in CD4+ T cells. Naive, effector memory (EM) reexpressing CD45RA T cells and follicular CD4+ T cells, as well as TRECs and HLA-DR+ and CD38+CD4+ activated T lymphocytes, were increased in those patients. EM2 and EM3 CD4+ T cells, EM2 CD8+ T cells and memory B cells were elevated in the COVID-19 CPP group. Numbers of neutrophils, monocytes and natural killer cells were equivalent but with increased activation in the recovered patients. CONCLUSIONS In the short-term following infection, COVID-19-recovered patients show persistent activation profiles in phagocytes, T-cell subtypes and natural killer cells. Meanwhile, increased production of lymphocytes, TRECs and naive T cells suggests immune response recovery, even in immunosuppressed patients and children with comorbidities. The clinical implications of these findings should be further studied.
Collapse
Affiliation(s)
- Vera Bain
- From the Departamento de Pediatria, Faculdade de Medicina FMUSP
| | | | - Olivia M Matsuo
- From the Departamento de Pediatria, Faculdade de Medicina FMUSP
| | | | - Yingying Zheng
- From the Departamento de Pediatria, Faculdade de Medicina FMUSP
| | - Andreia Rangel-Santos
- Laboratorio de Pediatria Clinica (LIM-36), Departamento de Pediatria, Hospital das Clinicas HCFMUSP
| | | | | | | | | | | | | | | | - Camilla Astley
- Applied Physiology and Nutrition Research Group-Center of Lifestyle Medicine, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil
| | | | | | | | | | - Patricia Palmeira
- Laboratorio de Pediatria Clinica (LIM-36), Departamento de Pediatria, Hospital das Clinicas HCFMUSP
| | | |
Collapse
|
3
|
Ghimire R, Shrestha R, Amaradhi R, Liu L, More S, Ganesh T, Ford AK, Channappanavar R. Toll-like receptor 7 (TLR7)-mediated antiviral response protects mice from lethal SARS-CoV-2 infection. J Virol 2025; 99:e0166824. [PMID: 40162785 PMCID: PMC12090760 DOI: 10.1128/jvi.01668-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced impaired antiviral immunity and excessive inflammatory responses cause lethal pneumonia. However, the in vivo roles of key pattern recognition receptors that elicit protective antiviral and fatal inflammatory responses, specifically in the lungs, are not well described. Coronaviruses possess single-stranded RNA genome that activates TLR7/8 to induce an antiviral interferon (IFN) and robust inflammatory cytokine response. Here, using wild-type and TLR7-deficient (TLR7-/-) mice infected with mouse-adapted SARS-CoV-2 (MA-CoV-2), we examined the role of TLR7 in the lung antiviral and inflammatory response and severe pneumonia. We showed that TLR7 deficiency significantly increased lung virus loads and morbidity/mortality, which correlated with reduced levels of type I IFNs (Ifna/b), type III IFNs (Ifnl), and IFN-stimulated genes (ISGs) in the lungs. A detailed evaluation of MA-CoV-2-infected lungs revealed increased neutrophil accumulation and lung pathology in TLR7-/- mice. We further showed that blocking type I IFN receptor (IFNAR) signaling enhanced SARS-CoV-2 replication in the lungs and caused severe lung pathology, leading to 100% mortality compared to infected control mice. Moreover, immunohistochemical assessment of the lungs revealed increased numbers of SARS-CoV-2 antigen-positive macrophages, pneumocytes, and bronchial epithelial cells in TLR7-/- and IFNAR-deficient mice compared to control mice. In summary, we conclusively demonstrated that despite TLR7-induced robust lung inflammation, TLR7-induced IFN/ISG responses suppress lung virus replication and pathology and provide protection against SARS-CoV-2-induced fatal pneumonia. Additionally, given the similar disease outcomes in control, TLR7-/-, and IFNAR-deficient MA-CoV-2-infected mice and coronavirus disease 2019 (COVID-19) patients, we propose that MA-CoV-2-infected mice constitute an excellent model for studying COVID-19.IMPORTANCESevere coronavirus disease 2019 (COVID-19) is caused by a delicate balance between a strong antiviral and an exuberant inflammatory response. A robust antiviral immunity and regulated inflammation are protective, while a weak antiviral response and excessive inflammation are detrimental. However, the key host immune sensors that elicit protective antiviral and inflammatory responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge are poorly defined. Here, we examined the role of viral RNA-mediated TLR7 activation in the lung antiviral and inflammatory responses in SARS-CoV-2-infected mice. We demonstrate that TLR7 deficiency led to a high rate of morbidity and mortality, which correlated with an impaired antiviral interferon (IFN)-I/III response, enhanced lung virus replication, and severe lung pathology. Furthermore, we show that blocking IFN-I signaling using anti-IFN receptor antibody promoted SARS-CoV-2 replication in the lungs and caused severe disease. These results provide conclusive evidence that TLR7 and IFN-I receptor deficiencies lead to severe disease in mice, replicating clinical features observed in COVID-19 patients.
Collapse
Affiliation(s)
- Roshan Ghimire
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Rakshya Shrestha
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Radhika Amaradhi
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lin Liu
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Sunil More
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alexandra K. Ford
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Rudragouda Channappanavar
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
4
|
Ha MK, Postovskaya A, Kuznetsova M, Meysman P, Van Deuren V, Van Ierssel S, De Reu H, Schippers J, Peeters K, Besbassi H, Heyndrickx L, Willems B, Mariën J, Bartholomeus E, Vercauteren K, Beutels P, Van Damme P, Lion E, Vlieghe E, Laukens K, Coenen S, Naesens R, Ariën KK, Ogunjimi B. Celluloepidemiology-A paradigm for quantifying infectious disease dynamics on a population level. SCIENCE ADVANCES 2025; 11:eadt2926. [PMID: 40378227 DOI: 10.1126/sciadv.adt2926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/15/2025] [Indexed: 05/18/2025]
Abstract
To complement serology as a tool in public health interventions, we introduced the "celluloepidemiology" paradigm where we leveraged pathogen-specific T cell responses at a population level to advance our epidemiological understanding of infectious diseases, using SARS-CoV-2 as a model. Applying flow cytometry and machine learning on data from more than 500 individuals, we showed that the number of T cells with positive expression of functional markers not only could distinguish patients who recovered from COVID-19 from controls and pre-COVID donors but also identify previously unrecognized asymptomatic patients from mild, moderate, and severe recovered patients. The celluloepidemiology approach was uniquely capable to differentiate health care worker groups with different SARS-CoV-2 exposures from each other. T cell receptor (TCR) profiling strengthened our analysis by revealing that SARS-CoV-2-specific TCRs were more abundant in patients than in controls. We believe that adding data on T cell reactivity will complement serology and augment the value of infection morbidity modeling for populations.
Collapse
Affiliation(s)
- My K Ha
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
| | - Anna Postovskaya
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- ADReM Data Lab, Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (biomina), University of Antwerp, Antwerp, Belgium
- Clinical Virology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Maria Kuznetsova
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), 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 Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (biomina), University of Antwerp, Antwerp, Belgium
| | - Vincent Van Deuren
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- ADReM Data Lab, Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (biomina), University of Antwerp, Antwerp, Belgium
| | - Sabrina Van Ierssel
- Department of General Internal Medicine, Infectious Disease and Tropical Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology (LEH), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Flow Cytometry and Cell Sorting Core Facility (FACSUA), University of Antwerp, Wilrijk, Belgium
| | - Jolien Schippers
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
| | - Karin Peeters
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Hajar Besbassi
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
| | - Leo Heyndrickx
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Betty Willems
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Joachim Mariën
- Department of Ecology and Evolutionary Biology, University of Antwerp, Antwerp, Belgium
- The Virus Ecology Group, Institute of Tropical Medicine, Antwerp, Belgium
| | - Esther Bartholomeus
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
| | - Koen Vercauteren
- Clinical Virology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Philippe Beutels
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Pierre Van Damme
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Centre for the Evaluation of Vaccination (CEV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Eva Lion
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Laboratory of Experimental Hematology (LEH), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Flow Cytometry and Cell Sorting Core Facility (FACSUA), University of Antwerp, Wilrijk, Belgium
| | - Erika Vlieghe
- Department of General Internal Medicine, Infectious Disease and Tropical Medicine, Antwerp University Hospital, Edegem, Belgium
- Global Health Institute, University of Antwerp, Wilrijk, 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 Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (biomina), University of Antwerp, Antwerp, Belgium
| | - Samuel Coenen
- Laboratory of Medical Microbiology (LMM), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
- Center for General Practice, Department of Family Medicine and Population Health (FAMPOP), University of Antwerp, Wilrijk, Belgium
| | - Reinout Naesens
- Department of Clinical Biology, Antwerp Hospital Network, Antwerp, Belgium
| | - Kevin K Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Benson Ogunjimi
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Department of Pediatrics, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
5
|
Pekayvaz K, Kilani B, Joppich M, Eivers L, Brambs S, Knottenberg V, Akgöl S, Yue K, Li L, Martinez-Navarro A, Kaiser R, Meißner N, Schulz H, Belz L, Akhalkatsi A, Stockhausen S, Mueller TT, Millonig S, Hartelt L, Gold C, Janjic A, Polewka V, Wendler F, Droste Zu Senden A, Titova A, Leunig A, Voelkl M, Engelmann B, Hernandez Petzsche MR, Boeckh-Behrens T, Liebig T, Winning S, Fandrey J, Dichgans M, Enard W, Zimmer R, Tiedt S, Massberg S, Nicolai L, Stark K. Immunothrombolytic monocyte-neutrophil axes dominate the single-cell landscape of human thrombosis and correlate with thrombus resolution. Immunity 2025; 58:1343-1358.e13. [PMID: 40280129 DOI: 10.1016/j.immuni.2025.03.020] [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: 08/30/2024] [Revised: 12/31/2024] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Thrombotic diseases remain the major cause of death and disability worldwide, and the contribution of inflammation is increasingly recognized. Thromboinflammation has been identified as a key pathomechanism, but an unsupervised map of immune-cell states, trajectories, and intercommunication at a single-cell level has been lacking. Here, we reveal innate leukocyte substates with prominent thrombolytic properties by employing single-cell omics measures on human stroke thrombi. Using in vivo and in vitro thrombosis models, we propose a pro-resolving monocyte-neutrophil axis, combining two properties: (1) NR4A1hi non-classical monocytes acquire a thrombolytic and neutrophil-chemoattractive phenotype, and (2) blood neutrophils are thereby continuously recruited to established thrombi through CXCL8-CXCR1 and CXCR2 and adopt a hypoxia-induced thrombus-resolving urokinase receptor (PLAUR)+ phenotype. This immunothrombolytic axis results in thrombus resolution. Together, with this immune landscape of thrombosis, we provide a valuable resource and introduce the concept of "immunothrombolysis" with broad mechanistic and translational implications at the crossroad of inflammation and thrombosis.
Collapse
Affiliation(s)
- Kami Pekayvaz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Badr Kilani
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Markus Joppich
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Luke Eivers
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Sophia Brambs
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sezer Akgöl
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Keyang Yue
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lukas Li
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Rainer Kaiser
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Nina Meißner
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Heiko Schulz
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Larissa Belz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sven Stockhausen
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Tonina T Mueller
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Simon Millonig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lea Hartelt
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Christoph Gold
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Aleksandar Janjic
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Vivien Polewka
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Franziska Wendler
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Anna Titova
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Alexander Leunig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Voelkl
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany; Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Bernd Engelmann
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Moritz R Hernandez Petzsche
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Tobias Boeckh-Behrens
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas Liebig
- Institute for Diagnostic and Interventional Neuroradiology, University Hospital, LMU Munich, Munich, Germany
| | - Sandra Winning
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Joachim Fandrey
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Martin Dichgans
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ralf Zimmer
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Massberg
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Leo Nicolai
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Konstantin Stark
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
6
|
Sedaghat-Rostami E, Carr BV, Yang L, Keep S, Lean FZX, Atkinson I, Fones A, Paudyal B, Kirk J, Vatzia E, Gubbins S, Bickerton E, Briggs E, Núñez A, McNee A, Moffat K, Freimanis G, Rollier C, Muir A, Richard AC, Angelopoulos N, Gerner W, Tchilian E. Porcine respiratory coronavirus as a model for acute respiratory disease: mechanisms of different infection outcomes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf066. [PMID: 40304579 DOI: 10.1093/jimmun/vkaf066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/11/2025] [Indexed: 05/02/2025]
Abstract
Porcine respiratory coronavirus (PRCV) is a naturally occurring pneumotropic coronavirus in the pig, providing a valuable large animal model to study acute respiratory disease. PRCV pathogenesis and the resulting immune response were investigated in pigs, the natural large animal host. We compared 2 strains, ISU-1 and 135, which induced differing levels of pathology in the respiratory tract to elucidate the mechanisms leading to mild or severe disease. The 135 strain induced greater pathology which was associated with higher viral load and stronger spike-specific antibody and T-cell responses. In contrast, the ISU-1 strain triggered mild pathology with a more balanced immune response and greater abundance of T regulatory cells. A higher frequency of putative T follicular helper cells was observed in animals infected with strain 135 at 11 days postinfection. Single-cell RNA-sequencing of bronchoalveolar lavage revealed differential gene expression in B and T cells between animals infected with 135 and ISU-1 at 1 day postinfection. These genes were associated with cell adhesion, migration, and immune regulation. Along with increased IL-6 and IL-12 production, these data indicate that heightened inflammatory responses to the 135 strain may contribute to pronounced pneumonia. Among bronchoalveolar lavage (BAL) immune cell populations, B cells and plasma cells exhibited the most gene expression divergence between pigs infected with different PRCV strains, highlighting their role in maintaining immune homeostasis in the respiratory tract. These findings indicate the potential of the PRCV model for studying coronavirus-induced respiratory disease and identifying mechanisms that determine infection outcomes.
Collapse
Affiliation(s)
- Ehsan Sedaghat-Rostami
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guilford, United Kingdom
| | | | - Liu Yang
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Sarah Keep
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Fabian Z X Lean
- Department of Pathology, Animal and Plant Health Agency, Addlestone, United Kingdom
| | - Isabella Atkinson
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Albert Fones
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Basudev Paudyal
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - James Kirk
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Eleni Vatzia
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Simon Gubbins
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Erica Bickerton
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Emily Briggs
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guilford, United Kingdom
| | - Alejandro Núñez
- Department of Pathology, Animal and Plant Health Agency, Addlestone, United Kingdom
| | - Adam McNee
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Katy Moffat
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Graham Freimanis
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Christine Rollier
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guilford, United Kingdom
| | - Andrew Muir
- Department of Pathology, Animal and Plant Health Agency, Addlestone, United Kingdom
| | | | - Nicos Angelopoulos
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Wilhelm Gerner
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Elma Tchilian
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| |
Collapse
|
7
|
Zhao Y, Xiong C, Wang B, Li D, Liu J, Wei S, Hou Y, Zhou Y, Zheng R. The Discovery of Phages in the Substantia Nigra and Its Implication for Parkinson's Disease. RESEARCH (WASHINGTON, D.C.) 2025; 8:0657. [PMID: 40308709 PMCID: PMC12041648 DOI: 10.34133/research.0657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/28/2025] [Accepted: 03/10/2025] [Indexed: 05/02/2025]
Abstract
Background: A century ago, a mystery between a virus and Parkinson's disease (PD) was described. Owing to the limitation of human brain biopsy and the challenge of electron microscopy in observing virions in human brain tissue, it has been difficult to study the viral etiology of PD. Recent discovery of virobiota reveals that viruses coexist with humans as symbionts. Newly developed transcriptomic sequencing and novel bioinformatic approaches for mining the encrypted virome in human transcriptome make it possible to study the relationship between symbiotic viruses and PD. Nevertheless, whether viruses exist in the human substantia nigra (SN) and whether symbiotic viruses underlie PD pathogenesis remain unknown. Methods: We collected current worldwide human SN transcriptomic datasets from the United States, the United Kingdom, the Netherlands, and Switzerland. We used bioinformatic approaches including viruSITE and the Viral-Track to identify the existence of viruses in the SN of patients. The comprehensive RNA sequencing-based virome analysis pipeline was used to characterize the virobiota in the SN. The Pearson's correlation analysis was used to examine the association between the viral RNA fragment counts (VRFCs) and PD-related human gene sequencing reads in the SN. The differentially expressed genes (DEGs) in the SN between PD patients and non-PD individuals were used to examine the molecular signatures of PD and also evaluate the impact of symbiotic viruses on the SN. Findings: We observed the existence of viruses in the human SN. A dysbiosis of virobiota was found in the SN of PD patients. A marked correlation between VRFC and PD-related human gene expression was detected in the SN of PD patients. These PD-related human genes correlated to VRFC were named as the virus-correlated PD-related genes (VPGs). We identified 3 bacteriophages (phages), including the Proteus phage VB_PmiS-Isfahan, the Escherichia phage phiX174, and the Lactobacillus phage Sha1, that might impair the gene expression of neural cells in the SN of PD patients. The Proteus phage VB_PmiS-Isfahan was a common virus in the SN of patients from the United Kingdom, the Netherlands, and Switzerland. VPGs and DEGs together highlighted that the phages might dampen dopamine biosynthesis and weaken the cGAS-STING function. Interpretation: This is the first study to discover the involvement of phages in PD pathogenesis. A lifelong low symbiotic viral load in the SN may be a contributor to PD pathogenesis. Our findings unlocked the black box between brain virobiota and PD, providing a novel insight into PD etiology from the perspective of phage-human symbiosis.
Collapse
Affiliation(s)
- Yun Zhao
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Changxian Xiong
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Bingwei Wang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Daotong Li
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Jiarui Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Shizhang Wei
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Yujia Hou
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
- Neuroscience Research Institute,
Peking University, Beijing, China
- Key Laboratory for Neuroscience of Ministry of Education,
Peking University, Beijing, China
- Key Laboratory for Neuroscience of National Health Commission,
Peking University, Beijing, China
- Beijing Life Science Academy, Beijing, China
| |
Collapse
|
8
|
Karl V, Hofmann M, Thimme R. Role of antiviral CD8+ T cell immunity to SARS-CoV-2 infection and vaccination. J Virol 2025; 99:e0135024. [PMID: 40029063 PMCID: PMC11998524 DOI: 10.1128/jvi.01350-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
The COVID-19 pandemic has greatly enhanced our understanding of CD8+ T cell immunity and their role in natural infection and vaccine-induced protection. Rapid and early SARS-CoV-2-specific CD8+ T cell responses have been associated with efficient viral clearance and mild disease. Virus-specific CD8+ T cell responses can compensate for waning, morbidity-related, and iatrogenic reduction of humoral immunity. After infection or vaccination, SARS-CoV-2-specific memory CD8+ T cells are formed, which mount an efficient recall response in the event of breakthrough infection and help to protect from severe disease. Due to their breadth and ability to target mainly highly conserved epitopes, SARS-CoV-2-specific CD8+ T cells are also able to cross-recognize epitopes of viral variants, thus maintaining immunity even after the emergence of viral evolution. In some cases, however, CD8+ T cells may contribute to the pathogenesis of severe COVID-19. In particular, delayed and uncontrolled, e.g., nonspecific and hyperactivated, cytotoxic CD8+ T cell responses have been linked to poor COVID-19 outcomes. In this minireview, we summarize the tremendous knowledge about CD8+ T cell responses to SARS-CoV-2 infection and COVID-19 vaccination that has been gained over the past 5 years, while also highlighting the critical knowledge gaps that remain.
Collapse
Affiliation(s)
- Vivien Karl
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Maike Hofmann
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Thimme
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
9
|
Besson J, Audran R, Karlen M, Miauton A, Hajjami HME, Warpelin-Decrausaz L, Sene L, Schaufelberger S, Faivre V, Faouzi M, Hartley MA, Spertini F, Genton B. A gold nanoparticle/peptide vaccine designed to induce SARS-CoV-2-specific CD8 T cells: a double-blind, randomized, phase 1 study in Switzerland. BMC Infect Dis 2025; 25:472. [PMID: 40197245 PMCID: PMC11974014 DOI: 10.1186/s12879-025-10844-3] [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: 10/21/2024] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND New vaccines with broader protection against SARS-CoV-2 are needed to reduce the risk of immune escape and provide broad and long-lasting cellular immunity. The objectives of the naNO-COVID trial were to evaluate the safety and immunogenicity of a CD8 + T cell, gold nanoparticle-based, peptide COVID-19 vaccine. METHODS A randomized, double-blind, vehicle-controlled, phase 1 trial in healthy adults to receive PepGNP-Covid19 or Vehicle-GNP, followed over 180 days, using a dose-escalation strategy. RESULTS Twenty participants received PepGNP-Covid19 (low dose [LD] or high dose [HD], n = 10 each) and six Vehicle-GNP (LD or HD, n = 3 each). Vaccinations were safe. No serious adverse events were reported. Most of the adverse events were mild, two adverse events of special interest related to the product (fever and fatigue). Reactogenicity was similar overall between vaccine, comparator, and doses. Virus-specific humoral responses in LD PepGNP-Covid19 and Vehicle-GNP groups coincided with SARS-CoV-2 infections. PepGNP-Covid19 vaccination induced the modulation of Covid19-specific CD137 + CD69 + CD8 + , and an increase at day 35 particularly in central and effector memory T cells in LD group, and in late effector memory cells in HD group. CONCLUSIONS The favourable safety profile and cellular responses observed support further development of PepGNP-Covid19. TRIAL REGISTRATION ClinicalTrials.gov, NCT05113862, approved 09.11.2021.
Collapse
Affiliation(s)
- Juliette Besson
- Tropical, Travel and Vaccination Clinic, Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland.
| | - Régine Audran
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Maxime Karlen
- Tropical, Travel and Vaccination Clinic, Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Alix Miauton
- Tropical, Travel and Vaccination Clinic, Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Hélène Maby-El Hajjami
- Clinical Trial Unit, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Loane Warpelin-Decrausaz
- Clinical Trial Unit, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
- Research Support Unit, Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Loredana Sene
- Clinical Trial Unit, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Sylvain Schaufelberger
- Information Systems and Digital Transformation, Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Vincent Faivre
- Information Systems and Digital Transformation, Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Mohamed Faouzi
- Division of Biostatistics, Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Mary-Anne Hartley
- Tropical, Travel and Vaccination Clinic, Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - François Spertini
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Blaise Genton
- Tropical, Travel and Vaccination Clinic, Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
10
|
Yang OO. The immunopathogenesis of SARS-CoV-2 infection: Overview of lessons learned in the first 5 years. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf033. [PMID: 40180332 DOI: 10.1093/jimmun/vkaf033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/11/2025] [Indexed: 04/05/2025]
Abstract
This review provides a broad overview of lessons learned in the five years since COVID-19 was identified. It is a bimodal disease, starting with an initially virus-driven phase, followed by resolution or ensuing inappropriate immune activation causing severe inflammation that is no longer strictly virus dependent. Humoral immunity is beneficial for preventing or attenuating the early stage, without benefit once the later stage begins. Neutralizing antibodies elicited by natural infection or vaccination are short-lived and highly vulnerable to viral sequence variation. By contrast, cellular immunity, particularly the CD8+ T cell arm, has a role in preventing or attenuating severe disease, is far less susceptible to viral variation, and is longer-lived than antibodies. Finally, an ill-defined phenomenon of prolonged symptoms after acute infection, termed "long COVID," is poorly understood but may involve various immunologic defects that are hyperactivating or immunosuppressive. Remaining issues include needing to better understand the immune dysregulation of severe disease to allow more tailored therapeutic interventions, developing antibody strategies that cope with the viral spike sequence variability, prolonging vaccine efficacy, and unraveling the mechanisms of long COVID to design therapeutic approaches.
Collapse
Affiliation(s)
- Otto O Yang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
Sawano M, Wu Y, Shah RM, Zhou T, Arun AS, Khosla P, Kaleem S, Vashist A, Bhattacharjee B, Ding Q, Lu Y, Caraballo C, Warner F, Huang C, Herrin J, Putrino D, Michelsen T, Fisher L, Adinig C, Iwasaki A, Krumholz HM. Long COVID Characteristics and Experience: A Descriptive Study From the Yale LISTEN Research Cohort. Am J Med 2025; 138:712-720.e13. [PMID: 38663793 DOI: 10.1016/j.amjmed.2024.04.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND The experience of people with long COVID needs further amplification, especially with a comprehensive focus on symptomatology, treatments, and the impact on daily life and finances. Our intent is to describe the experience of people with long COVID symptomatology and characterize the psychological, social, and financial challenges they experience. METHODS We collected data from individuals aged 18 and older reporting long COVID as participants in the Yale Listen to Immune, Symptom and Treatment Experiences Now study. The sample population included 441 participants surveyed between May 2022 and July 2023. We evaluated their demographic characteristics, socioeconomic and psychological status, index infection period, health status, quality of life, symptoms, treatments, prepandemic comorbidities, and new-onset conditions. RESULTS Overall, the median age of the participants with long COVID was 46 years (interquartile range [IQR]: 38-57 years); 74% were women, 86% were non-Hispanic White, and 93% were from the United States. Participants reported a low health status measured by the Euro-QoL visual analog scale, with a median score of 49 (IQR: 32-61). Participants documented a diverse range of symptoms, with all 96 possible symptom choices being reported. Additionally, participants had tried many treatments (median number of treatments: 19, IQR: 12-28). They were also experiencing psychological distress, social isolation, and financial stress. CONCLUSIONS Despite having tried numerous treatments, participants with long COVID continued to experience an array of health and financial challenges-findings that underscore the failure of the healthcare system to address the medical needs of people with long COVID. These insights highlight the need for crucial medical, mental health, financial, and community support services, as well as further scientific investigation to address the complex impact of long COVID.
Collapse
Affiliation(s)
- Mitsuaki Sawano
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Conn; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Yilun Wu
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Conn; Department of Biostatistics, Yale School of Public Health, New Haven, Conn
| | - Rishi M Shah
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Conn; Department of Applied Mathematics, Yale College, New Haven, Conn
| | | | | | | | - Shayaan Kaleem
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anushree Vashist
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Conn; The College at the University of Chicago, Chicago, Ill
| | - Bornali Bhattacharjee
- Center for Infection and Immunity, Yale School of Medicine, New Haven, Conn; Department of Immunobiology, Yale School of Medicine, New Haven, Conn
| | - Qinglan Ding
- College of Health and Human Sciences, Purdue University, West Lafayette, Ind
| | - Yuan Lu
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Conn; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn; Department of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, Conn; Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Conn
| | - César Caraballo
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Conn; Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Frederick Warner
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Conn; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Chenxi Huang
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Conn; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Jeph Herrin
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - David Putrino
- Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | | | - Akiko Iwasaki
- Center for Infection and Immunity, Yale School of Medicine, New Haven, Conn; Department of Immunobiology, Yale School of Medicine, New Haven, Conn; Howard Hughes Medical Institute, Chevy Chase, Md
| | - Harlan M Krumholz
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Conn; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn; Center for Infection and Immunity, Yale School of Medicine, New Haven, Conn; Department of Health Policy and Management, Yale School of Public Health, New Haven, Conn.
| |
Collapse
|
12
|
Cañón-Estrada F, Muñoz-Ordoñez JA, Escalante-Forero M, Rodas Y, Arteaga-Tobar AA, Azcarate-Rodriguez V, Perna E, Mendoza I, Wyss F, Barisani JL, Speranza M, Alarco W, Ortega JC, Ulate A, Mercedes J, Chaves DQ, Oliver P, Valencia-Orozco A, Barbosa MM, León-Giraldo H, Flórez NA, Gómez-Mesa JE. Biochemical differences based on sex and clusters of biomarkers in patients with COVID-19: analysis from the CARDIO COVID 19-20 registry. BMC Cardiovasc Disord 2025; 25:147. [PMID: 40045210 PMCID: PMC11881352 DOI: 10.1186/s12872-025-04565-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/10/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The inflammatory response associated with COVID-19 varies with sex, potentially affecting disease outcomes. Males have a higher risk of complications compared to females, requiring an evaluation of differences in inflammatory response severity based on sex. OBJECTIVE To compare clinical data, biochemical biomarkers, and outcomes among hospitalized COVID-19 patients in Latin America and the Caribbean (LA&C) based on sex and to perform a cluster analysis of biomarker profiles for both sexes. METHODS This prospective, multicenter observational registry made by the Inter-American Council of Heart Failure and Pulmonary Hypertension of the Inter-American Society of Cardiology included hospitalized COVID-19 patients from 44 hospitals in 14 countries in LA&C between May 1, 2020, and June 30, 2021. RESULTS Of 3,260 patients (1,201 females and 2,059 males), males had higher C-reactive protein and ferritin levels, while females had higher natriuretic peptides and d-dimer levels. Males had more cardiovascular complications (acute coronary syndrome [3.3% vs. 2.2%], decompensated heart failure [8.9% vs. 7.8%], pulmonary embolism [4.4% vs. 2.9%]), intensive care unit (ICU) admissions (56.9% vs. 47.7%), and overall mortality (27.5% vs. 22.1%). Cluster analysis identified three groups: one with normal-range biomarkers but elevated ferritin, one with coagulation abnormalities, and one with an inflammatory profile linked to renal injury and increased non-cardiovascular mortality. CONCLUSIONS In the LA&C population hospitalized with COVID-19, males had higher inflammatory biomarker levels, correlating with increased cardiovascular complications and mortality. The cluster with an inflammatory profile showed higher non-cardiovascular mortality, while clusters with elevated ferritin levels were associated with increased ICU admissions.
Collapse
Affiliation(s)
| | | | | | - Yorlany Rodas
- Centro de Investigaciones Clínicas, Fundación Valle del Lili, 76003, Cali, Colombia
| | | | | | - Eduardo Perna
- Departamento de Cardiología, Instituto de Cardiología J.F Cabral, Corrientes, 3400, Argentina
| | - Iván Mendoza
- Facultad de Ciencias de La Salud, Universidad Central de Venezuela, Caracas, 1030, Venezuela
| | - Fernando Wyss
- Departamento de Cardiología, Servicios y Tecnología Cardiovascular de Guatemala S.A-Cardiosolutions, Guatemala City, 01010, Guatemala
| | - José Luis Barisani
- Departamento Cardiovascular, Hospital Presidente Perón, 1710, Buenos Aires, Argentina
| | - Mario Speranza
- Departamento de Cardiología, Hospital Clínica Bíblica, San José, 10104, Costa Rica
| | - Walter Alarco
- Departamento de Cardiología, Instituto Nacional Cardiovascular INCOR ESSALUD, Lima, 1507, Perú
| | - Juan Carlos Ortega
- Departamento de Cardiología, Hospital Universitario Erasmo Meoz, 540003, Cúcuta, Colombia
| | - Andrés Ulate
- Departamento de Cardiología, Hospital México, San José, 10101, Costa Rica
| | - Jessica Mercedes
- Departamento de Cardiología, Hospital Nacional San Rafael, Santa Tecla, 1502, El Salvador
| | - Daniel Quesada Chaves
- Departamento de Cardiología, Hospital San Vicente de Paúl, Heredia, 40101, Costa Rica
| | - Paola Oliver
- Departamento de Cardiología, Hospital Nacional Arzobispo Loayza, Lima, 15072, Perú
| | | | - Mario Miguel Barbosa
- Centro de Investigaciones Clínicas, Fundación Valle del Lili, 76003, Cali, Colombia
| | - Hoover León-Giraldo
- Centro de Investigaciones Clínicas, Fundación Valle del Lili, 76003, Cali, Colombia
| | - Noel Alberto Flórez
- Departamento de Cardiología, Fundación Valle del Lili, Street 98 N. 18-49, 76003, Cali, Colombia
| | - Juan Esteban Gómez-Mesa
- Facultad de Ciencias de La Salud, Universidad Icesi, 76003, Cali, Colombia.
- Departamento de Cardiología, Fundación Valle del Lili, Street 98 N. 18-49, 76003, Cali, Colombia.
- Centro de Investigaciones Clínicas, Fundación Valle del Lili, 76003, Cali, Colombia.
| |
Collapse
|
13
|
Chakraborty C, Bhattacharya M, Das A, Saha A. Regulation of miRNA in Cytokine Storm (CS) of COVID-19 and Other Viral Infection: An Exhaustive Review. Rev Med Virol 2025; 35:e70026. [PMID: 40032584 DOI: 10.1002/rmv.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/29/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
In the initial stage of the COVID-19 pandemic, high case fatality was noted. The case fatality during this was associated with the cytokine storm (CS) or cytokine storm syndrome (CSS). Sometimes, virus infections are due to the excessive secretion of pro-inflammatory cytokines, leading to cytokine storms, which might be directed to ARDS, multi-organ failure, and death. However, it was noted that several miRNAs are involved in regulating cytokines during SARS-CoV-2 and other viruses such as IFNs, ILs, GM-CSF, TNF, etc. The article spotlighted several miRNAs involved in regulating cytokines associated with the cytokine storm caused by SARS-CoV-2 and other viruses (influenza virus, MERS-CoV, SARS-CoV, dengue virus). Targeting those miRNAs might help in the discovery of novel therapeutics, considering CS or CSS associated with different virus infections.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| | | | - Arpita Das
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| | - Abinit Saha
- Deparment of Zoology, J.K. College, Purulia, India
| |
Collapse
|
14
|
Rubio-Casillas A, Redwan EM, Uversky VN. More antibodies are not always better: Fc effector functions play a critical role in SARS-CoV-2 infection and protection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:413-447. [PMID: 40246351 DOI: 10.1016/bs.pmbts.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Traditional vaccinology has primarily focused on neutralizing antibody titers as the main correlate of vaccine efficacy, often overlooking the multifaceted roles of antibody Fc effector functions in orchestrating protective immune responses. Fc-mediated immune responses play a pivotal role in immune modulation and pathogen clearance. Emerging evidence from natural infections and vaccine studies highlights the critical contribution of Fc effector functions in determining the quality and durability of immunity. This work explores the limitations of current vaccine evaluation paradigms that prioritize neutralization over Fc effector mechanisms. It also describes findings from a study showing an unexpected role for SARS-CoV-2 anti-spike antibodies: both convalescent plasma and patient-derived monoclonal antibodies (mAbs) lead to maximum phagocytic capacity by monocytes at low concentrations, whereas at higher concentrations the phagocytic capacity was reduced. Given that the severity of COVID-19 disease and antibody titers are strongly positively correlated, this work challenges the paradigm that high antibodies offer better protection against severe disease. It is proposed that humoral and cellular responses elicited by vaccination should never be higher than those produced by natural infection. By integrating antibody Fc effector functions into vaccine development, a paradigm shift is proposed that emphasizes synergic antibody responses. Such an approach could transform vaccine efficacy assessment, enhance protection against dangerous pathogens, and drive innovation in vaccine design.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Jalisco Health Services, Autlan, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco, Mexico.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg El-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
15
|
Zaslavsky ME, Craig E, Michuda JK, Sehgal N, Ram-Mohan N, Lee JY, Nguyen KD, Hoh RA, Pham TD, Röltgen K, Lam B, Parsons ES, Macwana SR, DeJager W, Drapeau EM, Roskin KM, Cunningham-Rundles C, Moody MA, Haynes BF, Goldman JD, Heath JR, Chinthrajah RS, Nadeau KC, Pinsky BA, Blish CA, Hensley SE, Jensen K, Meyer E, Balboni I, Utz PJ, Merrill JT, Guthridge JM, James JA, Yang S, Tibshirani R, Kundaje A, Boyd SD. Disease diagnostics using machine learning of B cell and T cell receptor sequences. Science 2025; 387:eadp2407. [PMID: 39977494 PMCID: PMC12061481 DOI: 10.1126/science.adp2407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 11/29/2024] [Indexed: 02/22/2025]
Abstract
Clinical diagnosis typically incorporates physical examination, patient history, various laboratory tests, and imaging studies but makes limited use of the human immune system's own record of antigen exposures encoded by receptors on B cells and T cells. We analyzed immune receptor datasets from 593 individuals to develop MAchine Learning for Immunological Diagnosis, an interpretive framework to screen for multiple illnesses simultaneously or precisely test for one condition. This approach detects specific infections, autoimmune disorders, vaccine responses, and disease severity differences. Human-interpretable features of the model recapitulate known immune responses to severe acute respiratory syndrome coronavirus 2, influenza, and human immunodeficiency virus, highlight antigen-specific receptors, and reveal distinct characteristics of systemic lupus erythematosus and type-1 diabetes autoreactivity. This analysis framework has broad potential for scientific and clinical interpretation of immune responses.
Collapse
MESH Headings
- Humans
- Autoimmune Diseases/diagnosis
- Autoimmune Diseases/immunology
- B-Lymphocytes/immunology
- COVID-19/diagnosis
- COVID-19/immunology
- Diabetes Mellitus, Type 1/diagnosis
- Diabetes Mellitus, Type 1/immunology
- HIV Infections/diagnosis
- HIV Infections/immunology
- Influenza, Human/diagnosis
- Influenza, Human/immunology
- Lupus Erythematosus, Systemic/diagnosis
- Lupus Erythematosus, Systemic/immunology
- Machine Learning
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- SARS-CoV-2/immunology
- Infections/diagnosis
- Infections/immunology
Collapse
Affiliation(s)
| | - Erin Craig
- Department of Biomedical Data Science, Stanford University; Stanford, CA, USA
| | - Jackson K. Michuda
- Department of Biomedical Data Science, Stanford University; Stanford, CA, USA
| | - Nidhi Sehgal
- Department of Genetics, Stanford University; Stanford, CA, USA
- Department of Pathology, Stanford University; Stanford, CA, USA
| | - Nikhil Ram-Mohan
- Department of Emergency Medicine, Stanford University; Stanford, CA, USA
| | - Ji-Yeun Lee
- Department of Pathology, Stanford University; Stanford, CA, USA
| | - Khoa D. Nguyen
- Department of Pathology, Stanford University; Stanford, CA, USA
| | - Ramona A. Hoh
- Department of Pathology, Stanford University; Stanford, CA, USA
| | - Tho D. Pham
- Department of Pathology, Stanford University; Stanford, CA, USA
- Stanford Blood Center; Stanford, CA, USA
| | - Katharina Röltgen
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute; Allschwil, Switzerland
- University of Basel; Basel, Switzerland
| | - Brandon Lam
- Department of Pathology, Stanford University; Stanford, CA, USA
| | - Ella S. Parsons
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University; Stanford, CA, USA
| | - Susan R. Macwana
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation; Oklahoma City, OK, USA
| | - Wade DeJager
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation; Oklahoma City, OK, USA
| | - Elizabeth M. Drapeau
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Krishna M. Roskin
- Department of Pediatrics, University of Cincinnati, College of Medicine; Cincinnati, OH, USA
- Divisions of Biomedical Informatics and Immunobiology, Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, USA
| | | | - M. Anthony Moody
- Department of Pediatrics, Duke University; Durham, NC, USA
- Duke Human Vaccine Institute, Duke University; Durham, NC, USA
- Department of Immunology, Duke University; Durham, NC, USA
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke University; Durham, NC, USA
- Department of Immunology, Duke University; Durham, NC, USA
- Department of Medicine, Duke University; Durham, NC, USA
| | - Jason D. Goldman
- Swedish Center for Research and Innovation, Swedish Medical Center; Seattle, WA, USA
- Division of Allergy and Infectious Diseases, University of Washington; Seattle, WA, USA
| | - James R. Heath
- Institute for Systems Biology; Seattle, WA, USA
- Department of Bioengineering, University of Washington; Seattle, WA, USA
| | - R. Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University; Stanford, CA, USA
| | - Kari C. Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health; Boston, MA, USA
- Division of Allergy and Inflammation, Beth Israel Deaconess Medical Center; Boston, MA, USA
| | - Benjamin A. Pinsky
- Department of Pathology, Stanford University; Stanford, CA, USA
- Department of Medicine, Stanford University; Stanford, CA, USA
| | | | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Kent Jensen
- Department of Medicine, Stanford University; Stanford, CA, USA
| | - Everett Meyer
- Department of Medicine, Stanford University; Stanford, CA, USA
| | - Imelda Balboni
- Department of Pediatrics, Stanford University; Stanford, CA, USA
| | - Paul J Utz
- Department of Medicine, Stanford University; Stanford, CA, USA
| | - Joan T. Merrill
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation; Oklahoma City, OK, USA
- Department of Medicine, Grossman School of Medicine, New York University; New York, NY, USA
- Lupus Foundation of America; Washington, DC, USA
| | - Joel M. Guthridge
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation; Oklahoma City, OK, USA
| | - Judith A. James
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation; Oklahoma City, OK, USA
| | - Samuel Yang
- Department of Emergency Medicine, Stanford University; Stanford, CA, USA
| | - Robert Tibshirani
- Department of Biomedical Data Science, Stanford University; Stanford, CA, USA
- Department of Statistics, Stanford University; Stanford, CA, USA
| | - Anshul Kundaje
- Department of Computer Science, Stanford University; Stanford, CA, USA
- Department of Genetics, Stanford University; Stanford, CA, USA
| | - Scott D. Boyd
- Department of Pathology, Stanford University; Stanford, CA, USA
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University; Stanford, CA, USA
| |
Collapse
|
16
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
17
|
Asaba CN, Bitazar R, Labonté P, Bukong TN. Bronchoalveolar lavage single-cell transcriptomics reveals immune dysregulations driving COVID-19 severity. PLoS One 2025; 20:e0309880. [PMID: 39928675 PMCID: PMC11809808 DOI: 10.1371/journal.pone.0309880] [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: 08/20/2024] [Accepted: 12/30/2024] [Indexed: 02/12/2025] Open
Abstract
The continuous threats posed by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the virus that causes COVID-19, including the emergence of potentially more infectious and deadly variants, necessitate ongoing studies to uncover novel and detailed mechanisms driving disease severity. Using single-cell transcriptomics, we conducted a secondary data analysis of bronchoalveolar lavage fluid (BALF) from COVID-19 patients of varying severities and healthy controls to comprehensively examine immune responses. We observed significant immune cell alterations correlating with disease severity. In severe cases, macrophages showed upregulation of pro-inflammatory genes TNFα and IL1β, contributing to severe inflammation and tissue damage. Neutrophils exhibited increased activation, marked by S100A8, CXCL8, and IL1β expression, with extended viability and reduced phagocytosis. Genes such as MCL1 and HIF1α supported extended viability, while MSR1 and MRC1 indicated reduced phagocytosis. Enhanced formation of neutrophil extracellular traps (NETs) and reduced clearance, indicated by NET-associated markers, were linked to thrombo-inflammation and organ damage. Both macrophages and neutrophils in severe cases showed impaired efferocytosis, indicated by decreased expression of MSR1 and TREM2 in macrophages and downregulation of FCGR3B in neutrophils, leading to the accumulation of apoptotic cells and exacerbating inflammation. Severe cases were characterized by M1 macrophages with high TNFα and IL1β, while milder cases had M2 macrophages with elevated PPARγ. Dendritic cells (DCs) in severe cases exhibited reduced proportions and attenuated expression of MHC class I genes (HLA-A, HLA-B, HLA-C) and co-stimulatory molecules (CD80, CD86), alongside increased cytochrome c expression, indicating impaired antigen presentation and enhanced apoptosis. NK and T cells in severe cases demonstrated altered receptor and gene expression, with increased activation markers IFNγ and ISG15, suggesting a paradoxical state of activation and exhaustion. This analysis highlights the critical role of dysregulated neutrophil, macrophage, dendritic cell, NK, and T cell responses in severe COVID-19, identifying potential therapeutic targets and providing novel insights into the disease.
Collapse
Affiliation(s)
- Clinton Njinju Asaba
- Armand-Frappier Santé Biotechnologie Research Center, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Razieh Bitazar
- Armand-Frappier Santé Biotechnologie Research Center, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Patrick Labonté
- Armand-Frappier Santé Biotechnologie Research Center, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Terence Ndonyi Bukong
- Armand-Frappier Santé Biotechnologie Research Center, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| |
Collapse
|
18
|
Shankar-Hari M, Francois B, Remy KE, Gutierrez C, Pastores S, Daix T, Jeannet R, Blood J, Walton AH, Salomao R, Auzinger G, Striker D, Martin RS, Anand NJ, Bosanquet J, Blood T, Brakenridge S, Moldawer LL, Vachharajani V, Yee C, Dal-Pizzol F, Morre M, Berbille F, van den Brink M, Hotchkiss R. A randomized, double-blind, placebo-controlled trial of IL-7 in critically ill patients with COVID-19. JCI Insight 2025; 10:e189150. [PMID: 39903535 PMCID: PMC11949036 DOI: 10.1172/jci.insight.189150] [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: 11/12/2024] [Accepted: 01/31/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Lymphopenia and failure of lymphocytes to mount an early IFN-γ response correlate with increased mortality in COVID-19. Given the essential role of CD4 helper and CD8 cytotoxic cells in eliminating viral pathogens, this profound loss in lymphocytes may impair patients' ability to eliminate the virus. IL-7 is a pleiotropic cytokine that is obligatory for lymphocyte survival and optimal function. METHODS We conducted a prospective, double-blind, randomized, placebo-controlled trial of CYT107, recombinant human IL-7, in 109 critically ill, patients with lymphopenia who have COVID-19. The primary endpoint was to assess CYT107's effect on lymphocyte recovery with secondary clinical endpoints including safety, ICU and hospital length-of-stay, incidence of secondary infections, and mortality. RESULTS CYT107 was well tolerated without precipitating a cytokine storm or worsening pulmonary function. Absolute lymphocyte counts increased in both groups without a significant difference between CYT107 and placebo. Patients with COVID-19 receiving CYT107 but not concomitant antiviral medications, known inducers of lymphopenia, had a final lymphocyte count that was 43% greater than placebo (P = 0.067). There were significantly fewer treatment-emergent adverse events in CYT107 versus placebo-treated patients (P < 0.001), consistent with a beneficial drug effect. Importantly, CYT107-treated patients had 44% fewer hospital-acquired infections versus placebo-treated patients (P = 0.014). CONCLUSION Given that hospital-acquired infections are responsible for a large percentage of COVID-19 deaths, this effect of CYT107 to decrease nosocomial infections could substantially reduce late morbidity and mortality in this highly lethal disease. The strong safety profile of CYT107 and its excellent tolerability provide support for trials of CYT107 in other potential pandemic respiratory viral infections. TRIAL REGISTRATION NCT04379076, NCT04426201, NCT04442178, NCT04407689, NCT04927169. FUNDING Funding for the trial was provided by RevImmune and the Cancer Research Institute.
Collapse
Affiliation(s)
- Manu Shankar-Hari
- Department of Translational Critical Care Medicine, Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Scotland, United Kingdom
| | - Bruno Francois
- Medical-Surgical ICU & Inserm CIC 1435 Centre Hospitalier Universitaire, Limoges, France
| | - Kenneth E. Remy
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Cristina Gutierrez
- Department of Critical Care Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephen Pastores
- Department of Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Thomas Daix
- Medical-Surgical ICU & Inserm CIC 1435 Centre Hospitalier Universitaire, Limoges, France
| | - Robin Jeannet
- Medical-Surgical ICU & Inserm CIC 1435 Centre Hospitalier Universitaire, Limoges, France
| | - Jane Blood
- Department of Anesthesiology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Andrew H. Walton
- Department of Anesthesiology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Reinaldo Salomao
- Department of Medicine, Universidade Federal de Sao Paulo (Unifesp), Sao Paulo, Brazil
| | - Georg Auzinger
- Department of Intensive Care Medicine, King’s College Hospital, London, United Kingdom
| | - David Striker
- Department of Critical Care Medicine, Missouri Baptist Medical Center, St. Louis, Missouri, USA
| | - Robert S. Martin
- Department of Critical Care Medicine, Missouri Baptist Medical Center, St. Louis, Missouri, USA
| | - Nitin J. Anand
- Department of Critical Care Medicine, Missouri Baptist Medical Center, St. Louis, Missouri, USA
| | - James Bosanquet
- Department of Critical Care Medicine, Missouri Baptist Medical Center, St. Louis, Missouri, USA
| | - Teresa Blood
- Department of Anesthesiology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Scott Brakenridge
- Department of Surgery, Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Lyle L. Moldawer
- Department of Surgery, Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Vidula Vachharajani
- Department of Critical Care Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Cassian Yee
- Department of Critical Care Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Felipe Dal-Pizzol
- Department of Medicine, Hospital Sao Jose, Criciuma, Santa Catarina, Brazil
| | | | | | | | - Richard Hotchkiss
- Department of Anesthesiology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
19
|
Chen H, Li Y, Yuan L, Liu F, Sun Q, Luo Q, Lei Y, Hou Y, Li J, Cai L, Tang S. Age-related immune response disparities between adults and children with severe COVID-19: a case-control study in China. Front Microbiol 2025; 16:1525051. [PMID: 39967737 PMCID: PMC11832681 DOI: 10.3389/fmicb.2025.1525051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Background Elucidation of immune response differences is critical for uncovering underlying mechanisms and developing potential intervention measures among adults and children with COVID-19. Methods In this retrospective study, we analyzed serum biochemical markers and cytokine profiles among adults and children with COVID-19 in the First People's Hospital of Chenzhou in Hunan, China from 1 December 2022 to 13 February 2023. A case-control study was conducted using propensity score matching (PSM) to mitigate possible confounding factors. Results The significant differences observed included lymphocyte exhaustion, an increased neutrophil-to-lymphocyte (NEU/LYM) ratio, high levels of C-reactive protein (CRP), and a cytokine storm, characterized by high levels of Th1 proinflammatory cytokines, including interleukin 1β (IL-1β), IL-6, IL-8, interferon type I (IFN-γ), and tumor necrosis factor (TNF-α) in the lung among severe adult COVID-19 patients. Additionally, systemic immune responses were observed in children with COVID-19. Conclusion Significant differences in immune responses between adults and children with COVID-19 highlight the different mechanisms and potential intervention measures of COVID-19.
Collapse
Affiliation(s)
- Hongliang Chen
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
- Department of Clinical Microbiology Laboratory, The First People's Hospital of Chenzhou, Chenzhou, China
| | - Yuan Li
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Liping Yuan
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fen Liu
- Department of Clinical Microbiology Laboratory, The First People's Hospital of Chenzhou, Chenzhou, China
| | - Qian Sun
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
- Wenzhou Center for Disease Control and Prevention, Wenzhou, Zhejiang, China
| | - Qingkai Luo
- Department of Clinical Microbiology Laboratory, The First People's Hospital of Chenzhou, Chenzhou, China
| | - Yefei Lei
- Department of Clinical Microbiology Laboratory, The First People's Hospital of Chenzhou, Chenzhou, China
| | - Yinglan Hou
- Department of Clinical Microbiology Laboratory, The First People's Hospital of Chenzhou, Chenzhou, China
| | - Jiayan Li
- Department of Clinical Microbiology Laboratory, The First People's Hospital of Chenzhou, Chenzhou, China
| | - Liang Cai
- Hunan Provincial Center for Disease Control and Prevention, Hunan, China
| | - Shixing Tang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Riemann L, Gutierrez R, Odak I, Barros-Martins J, Roesner LM, Leon Lara X, Falk C, Schulz TF, Hansen G, Werfel T, Förster R. Integrative deep immune profiling of the elderly reveals systems-level signatures of aging, sex, smoking, and clinical traits. EBioMedicine 2025; 112:105558. [PMID: 39862806 PMCID: PMC11873576 DOI: 10.1016/j.ebiom.2025.105558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/13/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Aging increases disease susceptibility and reduces vaccine responsiveness, highlighting the need to better understand the aging immune system and its clinical associations. Studying the human immune system, however, remains challenging due to its complexity and significant inter-individual variability. METHODS We conducted an immune profiling study of 550 elderly participants (≥60 years) and 100 young controls (20-40 years) from the RESIST Senior Individuals (SI) cohort. Extensive demographic, clinical, and laboratory data were collected. Multi-color spectral flow cytometry and 48-plex plasma cytokine assays were used for deep immune phenotyping. Data were analyzed using unsupervised clustering and multi-dataset integration approaches. FINDINGS We studied 97 innate and adaptive immune cell populations, revealing intricate age- and sex-related changes in the elderly immune system. Our large sample size allowed detection of even subtle changes in cytokines and immune cell clusters. Integrative analysis combining clinical, laboratory, and immunological data revealed systems-level aging signatures, including shifts in specific immune cell subpopulations and cytokine concentrations (e.g., HGF and CCL27). Additionally, we identified unique immune signatures associated with smoking, obesity, and diseases such as osteoporosis, heart failure, and gout. INTERPRETATION This study provides one of the most comprehensive immune profiles of elderly individuals, uncovering high-resolution immune changes associated with aging. Our findings highlight clinically relevant immune signatures that enhance our understanding of aging-related diseases and could guide future research into new treatments, offering translational insights into human health and aging. FUNDING Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany's Excellence Strategy-EXC 2155-project number 390874280.
Collapse
Affiliation(s)
- Lennart Riemann
- Institute of Immunology, Hannover Medical School, Hannover, Germany; Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.
| | | | - Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany; The Tisch Cancer Institute, Icahn School of Medicine, New York, USA
| | - Joana Barros-Martins
- Institute of Immunology, Hannover Medical School, Hannover, Germany; Department of Microbiology and Immunology, Columbia University Medical Center, New York, USA
| | - Lennart M Roesner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Ximena Leon Lara
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christine Falk
- Institute of Transplantation Immunology, Hannover Medical School, Hannover, Germany; German Centre for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Thomas F Schulz
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany; German Centre for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany; Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany; German Center of Lung Research (DZL), BREATH, Hannover, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany; German Centre for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany.
| |
Collapse
|
21
|
Eltayeb A, Redwan EM. T-cell immunobiology and cytokine storm of COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:1-30. [PMID: 40246342 DOI: 10.1016/bs.pmbts.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The 2019 coronavirus illness (COVID 2019) first manifests as a newly identified pneumonia and may quickly escalate to acute respiratory distress syndrome, which has caused a global pandemic. Except for individualized supportive care, no curative therapy has been steadfastly advised for COVID-19 up until this point. T cells and virus-specific T lymphocytes are required to guard against viral infection, particularly COVID-19. Delayed immunological reconstitution (IR) and cytokine storm (CS) continue to be significant barriers to COVID-19 cure. While severe COVID-19 patients who survived the disease had considerable lymphopenia and increased neutrophils, especially in the elderly, their T cell numbers gradually recovered. Exhausted T lymphocytes and elevated levels of pro-inflammatory cytokines, including IL6, IL10, IL2, and IL17, are observed in peripheral blood and the lungs. It implies that while convalescent plasma, IL-6 blocking, mesenchymal stem cells, and corticosteroids might decrease CS, Thymosin α1 and adaptive COVID-19-specific T cells could enhance IR. There is an urgent need for more clinical research in this area throughout the world to open the door to COVID-19 treatment in the future.
Collapse
Affiliation(s)
- Ahmed Eltayeb
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
22
|
Li L, Zhang X, Yan H, Dai M, Gao H, Wang Y, Jiang P, Dai E. Different immunological characteristics of asymptomatic and symptomatic COVID-19 patients without vaccination in the acute and convalescence stages. PeerJ 2025; 13:e18451. [PMID: 39897496 PMCID: PMC11786710 DOI: 10.7717/peerj.18451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 10/14/2024] [Indexed: 02/04/2025] Open
Abstract
The immune status of Coronavirus disease 2019 (COVID-19) patients in different stages of infection remains difficult to determine. In this study, we performed high-throughput single-cell mass cytometry on peripheral blood samples from 10 COVID-19 patients and four healthy donors to analyze their immune status at acute and convalescence phases. During the acute stage, the proportion of neutrophils increased significantly while natural killer (NK) cells decreased. In contrast, during the convalescence phase, the proportion of plasma cells decreased from the acute stage of disease onset and was lower than normal. The proportions of B, mast and plasma cell subsets decreased significantly with the process of disease recovery. Further analysis of the subsets of major immune cell types in COVID-19 patients with different clinical presentations in different stages showed that in the acute stages of disease progression, the T helper cell 1 (Th1), IgD+ B and neutrophil subsets increased in COVID-19 patients, especially in symptomatic patients, while the central memory CD4+T cells (CD4 TCM), mucosa-associated invariant T (MAIT) and NK cell subsets decreased significantly, especially in symptomatic patients. Then CD4 TCM and MAIT returned to normal levels at the recovery phase. Dynamic assessment displayed that the immune imbalance at the onset of COVID-19 could be corrected during recovery. Our study provides additional information on the immune status of COVID-19 patients with different clinical manifestations in different stages. These findings may provide new insights into COVID-19 immunotherapy and immune intervention.
Collapse
Affiliation(s)
- Li Li
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Intensive Care Unit, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Xin Zhang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Department of Tuberculosis, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Huimin Yan
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Clinical Research Center, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Muwei Dai
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University and Hebei Cancer Hospital, Shijiazhuang, Hebei, China
| | - Huixia Gao
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Department of Laboratory Medicine, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Yuling Wang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Department of Laboratory Medicine, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Ping Jiang
- Department of Cardiovascular Medicine, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Erhei Dai
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Department of Laboratory Medicine, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| |
Collapse
|
23
|
Hromić-Jahjefendić A, Aljabali AAA. Analysis of the immune response in COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:31-71. [PMID: 40246347 DOI: 10.1016/bs.pmbts.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The COVID-19 pandemic, instigated by the novel coronavirus SARS-CoV-2, has emerged as a significant global health challenge, demanding a profound grasp of the immune response. The innate immune system, a multifaceted network encompassing pattern recognition receptors (PRRs) and effector cells, assumes a pivotal function in detecting and countering this viral assailant. Toll-like receptors (TLRs), situated on immune cell surfaces and within endosomes, play a central role in recognizing SARS-CoV-2. TLR-2 and TLR-4 discern specific viral constituents, such as the spike (S) protein, setting off inflammatory signaling cascades and catalyzing the generation of type I interferons. Intracellular PRRs, including the RIG-I-like receptors (RLRs), RIG-I and MDA5, detect viral RNA within the cytoplasm of infected cells, provoking antiviral responses by initiating the synthesis of type I interferons. The equilibrium between interferons and pro-inflammatory cytokines dictates the outcomes of the disease. Interferons play an indispensable role in governing viral replication, while unregulated cytokine production can result in tissue harm and inflammation. This intricate dynamic underpins therapeutic strategies aimed at regulating immune responses in individuals grappling with COVID-19. Natural killer (NK) cells, with their capacity to recognize infected cells through the "missing self" phenomenon and activating receptors, make significant contributions to the defense against SARS-CoV-2. NK cells play a pivotal role in eliminating infected cells and boosting immune responses through antibody-dependent cell-mediated cytotoxicity (ADCC). In conclusion, comprehending the interplay among PRRs, interferons, and NK cells within innate immunity is paramount for discerning and combatting SARS-CoV-2. This comprehension illuminates therapeutic interventions and vaccine development, casting light on our endeavors to confront this worldwide health crisis.
Collapse
Affiliation(s)
- Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina.
| | - Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| |
Collapse
|
24
|
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
|
25
|
Laffranchi M, Paraboschi EM, Bianchetto-Aguilera F, Tamassia N, Gasperini S, Gardiman E, Piserà A, Del Prete A, Invernizzi P, Gismondi A, Mantovani A, Cassatella MA, Asselta R, Sozzani S. Neutrophils restricted contribution of CCRL2 genetic variants to COVID-19 severity. Heliyon 2025; 11:e41267. [PMID: 39811276 PMCID: PMC11731188 DOI: 10.1016/j.heliyon.2024.e41267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/12/2024] [Accepted: 12/14/2024] [Indexed: 01/02/2025] Open
Abstract
The 3p21.31 locus is the most robust genomic region associated with COVID-19 severity. This locus contains a main chemokine receptor (CKR) cluster. We tested expression quantitative trait loci (eQTL) targeting the 3p21.31 CKR cluster linked to COVID-19 hospitalization in Europeans from the COVID-19 HGI meta-analysis. Among these, CCRL2, a key regulator of neutrophil trafficking, was targeted by neutrophil-restricted eQTLs. We confirmed these eQTLs in an Italian COVID-19 cohort. Haplotype analysis revealed a link between an increased CCRL2 expression and COVID-19 severity and hospitalization. By the exposure of neutrophils to a TLR8 ligand, reflecting a viral infection, we revealed specific chromatin domains within the 3p21.31 locus exclusive to neutrophils. In addition, the identified variants mapped within these regions altered the binding motif of neutrophils-expressed transcription factors. These results support that CCRL2 eQTL variants contribute to the risk of severe COVID-19 by selectively affecting neutrophil functions.
Collapse
Affiliation(s)
- Mattia Laffranchi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Elvezia Maria Paraboschi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Sara Gasperini
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Elisa Gardiman
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Arianna Piserà
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), IRCCS Fondazione San Gerardo Dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Angela Gismondi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Marco A. Cassatella
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Silvano Sozzani
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
26
|
La Vecchia G, Del Buono MG, Bonaventura A, Vecchiè A, Moroni F, Sanna T, Abbate A. Inflammatory Heart Disease in Multisystem Inflammatory Syndrome. Curr Cardiol Rep 2025; 27:10. [PMID: 39775145 PMCID: PMC11711706 DOI: 10.1007/s11886-024-02173-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/31/2024] [Indexed: 01/11/2025]
Abstract
PURPOSEOF THE REVIEW In this review article, we aim to provide an overview of the pathophysiology, the clinical features, the therapeutic management and prognosis of patients affected by Multisystemic inflammatory syndrome (MIS) with cardiac involvement, focusing on myocarditis and pericarditis. RECENT FINDINGS MIS is a multiorgan hyperinflammatory condition due to a cytokine storm following (within 4-12 weeks) SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) infection. First described in children, it also affects young adults without comorbidities, predominantly males with highly heterogeneous clinical manifestations, including cardiac involvement. Pericardial and myocardial involvement are prevalent among patients affected by MIS leading to different clinical manifestations including myocarditis with arrhythmias, acute heart failure and cardiogenic shock that significantly affect the patient's prognosis. The heterogeneity of its clinical features and the significant overlap with other hyperinflammatory diseases make the diagnosis particularly challenging. Moreover, the evidence on the efficacy of pharmacological treatments targeting the hyperinflammatory response is scarce, as well as data on long-term prognosis.
Collapse
Affiliation(s)
- Giulia La Vecchia
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00128, Rome, Italy
- Center of Excellence in Cardiovascular Sciences, Isola Tiberina Hospital Gemelli Isola, Rome, Italy
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00128, Rome, Italy.
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 1, 00168, Rome, Italy.
| | - Aldo Bonaventura
- Ospedale Di Circolo E Fondazione Macchi, DepartmentofInternalMedicine, S.C. Medicina Generale 1, Medical Center, ASSTSetteLaghi, Varese, Italy
| | - Alessandra Vecchiè
- Ospedale Di Circolo E Fondazione Macchi, DepartmentofInternalMedicine, S.C. Medicina Generale 1, Medical Center, ASSTSetteLaghi, Varese, Italy
| | - Francesco Moroni
- Robert M. Berne Cardiovascular Research Center, and Department of Medicine, DivisionofCardiovascularMedicine,HeartandVascularCenter, University of Virginia, Charlottesville, VA, USA
| | - Tommaso Sanna
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00128, Rome, Italy
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, and Department of Medicine, DivisionofCardiovascularMedicine,HeartandVascularCenter, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
27
|
Tu TH, Bennani FE, Masroori N, Liu C, Nemati A, Rozza N, Grunbaum AM, Kremer R, Milhalcioiu C, Roy DC, Rudd CE. The identification of a SARs-CoV2 S2 protein derived peptide with super-antigen-like stimulatory properties on T-cells. Commun Biol 2025; 8:14. [PMID: 39762551 PMCID: PMC11704208 DOI: 10.1038/s42003-024-07350-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Severe COVID-19 can trigger a cytokine storm, leading to acute respiratory distress syndrome (ARDS) with similarities to superantigen-induced toxic shock syndrome. An outstanding question is whether SARS-CoV-2 protein sequences can directly induce inflammatory responses. In this study, we identify a region in the SARS-CoV-2 S2 spike protein with sequence homology to bacterial super-antigens (termed P3). Computational modeling predicts P3 binding to sites on MHC class I/II and the TCR that partially overlap with sites for the binding of staphylococcal enterotoxins B and H. Like SEB and SEH derived peptides, P3 stimulated 25-40% of human CD4+ and CD8 + T-cells, increasing IFN-γ and granzyme B production. viSNE and SPADE profiling identified overlapping and distinct IFN-γ+ and GZMB+ subsets. The super-antigenic properties of P3 were further evident by its selective expansion of T-cells expressing specific TCR Vα and Vβ chain repertoires. In vivo experiments in mice revealed that the administration of P3 led to a significant upregulation of proinflammatory cytokines IL-1β, IL-6, and TNF-α. While the clinical significance of P3 in COVID-19 remains unclear, its homology to other mammalian proteins suggests a potential role for this peptide family in human inflammation and autoimmunity.
Collapse
Affiliation(s)
- Thai Hien Tu
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Fatima Ezzahra Bennani
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Nasser Masroori
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Chen Liu
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Atena Nemati
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Nicholas Rozza
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Amichai Meir Grunbaum
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Richard Kremer
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Catalin Milhalcioiu
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
- Department of Medical Oncology, McGill University Health Center, Montreal, QC, Canada
| | - Denis-Claude Roy
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Christopher E Rudd
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada.
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada.
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada.
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco.
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada.
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada.
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
28
|
Adilović M, Hromić-Jahjefendić A, Mahmutović L, Šutković J, Rubio-Casillas A, Redwan EM, Uversky VN. Intrinsic Factors Behind the Long-COVID: V. Immunometabolic Disorders. J Cell Biochem 2025; 126:e30683. [PMID: 39639607 DOI: 10.1002/jcb.30683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/02/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
The complex link between COVID-19 and immunometabolic diseases demonstrates the important interaction between metabolic dysfunction and immunological response during viral infections. Severe COVID-19, defined by a hyperinflammatory state, is greatly impacted by underlying chronic illnesses aggravating the cytokine storm caused by increased levels of Pro-inflammatory cytokines. Metabolic reprogramming, including increased glycolysis and altered mitochondrial function, promotes viral replication and stimulates inflammatory cytokine production, contributing to illness severity. Mitochondrial metabolism abnormalities, strongly linked to various systemic illnesses, worsen metabolic dysfunction during and after the pandemic, increasing cardiovascular consequences. Long COVID-19, defined by chronic inflammation and immune dysregulation, poses continuous problems, highlighting the need for comprehensive therapy solutions that address both immunological and metabolic aspects. Understanding these relationships shows promise for effectively managing COVID-19 and its long-term repercussions, which is the focus of this review paper.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Lejla Mahmutović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Jasmin Šutković
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan, Mexico
- Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Mexico
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
29
|
Dieter RS, Kempaiah P, Dieter EG, Alcazar A, Tafur A, Gerotziafas G, Gonzalez Ochoa A, Abdesselem S, Biller J, Kipshidze N, Vandreden P, Guerrini M, Dieter RA, Durvasula R, Singh M, Fareed J. Cardiovascular Symposium on Perspectives in Long COVID. Clin Appl Thromb Hemost 2025; 31:10760296251319963. [PMID: 39943820 PMCID: PMC11822813 DOI: 10.1177/10760296251319963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/09/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Significant progress has been made in treating Coronavirus disease (COVID) - an infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). An ominous turn in the pandemic is the evolving public health crisis emanating from persistent SARS-CoV-2 infection and its associated long-term impact. Long COVID or post-COVID syndrome describes protean symptoms that persist at least 3 months after the onset of acute illness and last for at least 2 months in individuals with a history of confirmed SARS-CoV-2 infection. Long COVID has become a public health concern. Millions of infected individuals are now facing chronic multi-organ failures, including neuropsychiatric, cardiovascular, pulmonary, and kidney complications. In general, the cause of long COVID syndrome is unclear but factors such as prolonged activation of immune responses, and viral persistence triggering transcription dysregulation of genes associated with normal thrombotic disease may play a role in cardiovascular complications. Although inflammatory biomarkers are reported in other disorders, it remains unclear whether similar biomarkers are associated with cardiovascular manifestations following COVID. Medications such as sulodexide directed at glycocalyx and coagulation have demonstrated benefits for long COVID in smaller studies. Here, we describe the outcomes of the symposium on the underlying cardiovascular mechanisms of the long COVID.
Collapse
Affiliation(s)
- Robert S. Dieter
- Loyola University Stritch School of Medicine, Maywood, USA
- VA Hines, IL, USA
| | - Prakasha Kempaiah
- Loyola University Stritch School of Medicine, Maywood, USA
- Loyola University Chicago, Maywood, IL, USA
| | | | | | - Alfonso Tafur
- Endeavor Health, University of Chicago, Pritzker School of Medicine, Chicago, IL, USA
| | - Grigoris Gerotziafas
- Sorbonne University, INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team “Cancer, Vessels, Biology and Therapeutics” (CaVITE), Group “Cancer – Angiogenesis – Thrombosis”, University Institute of Cancerology (UIC), Saint Antoine University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
- Thrombosis and Haemostasis Center, Department of Obstetrics, Gynecology and Perinatal Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | | | - Jose Biller
- Loyola University Stritch School of Medicine, Maywood, USA
| | | | - Patrick Vandreden
- Sorbonne University, INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team “Cancer, Vessels, Biology and Therapeutics” (CaVITE), Group “Cancer – Angiogenesis – Thrombosis”, University Institute of Cancerology (UIC), Saint Antoine University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
- Department of Clinical Research, Diagnostica Stago, Gennevilliers, France
| | - Marco Guerrini
- Istituto di Ricerche Chimiche e Biochimiche G-Ronzoni – NMR Center, Milano, Italy
| | | | | | - Meharvan Singh
- Loyola University Stritch School of Medicine, Maywood, USA
- Loyola University Chicago, Maywood, IL, USA
| | - Jawed Fareed
- Loyola University Stritch School of Medicine, Maywood, USA
- Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
30
|
Nguema L, Picard F, El Hajj M, Dupaty L, Fenwick C, Cardinaud S, Wiedemann A, Pantaleo G, Zurawski S, Centlivre M, Zurawski G, Lévy Y, Godot V. Subunit protein CD40.SARS.CoV2 vaccine induces SARS-CoV-2-specific stem cell-like memory CD8 + T cells. EBioMedicine 2025; 111:105479. [PMID: 39667270 PMCID: PMC11697708 DOI: 10.1016/j.ebiom.2024.105479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Ideally, vaccination should induce protective long-lived humoral and cellular immunity. Current licensed COVID-19 mRNA vaccines focused on the spike (S) region induce neutralizing antibodies that rapidly wane. METHODS Herein, we show that a subunit vaccine (CD40.CoV2) targeting spike and nucleocapsid antigens to CD40-expressing cells elicits broad specific human (hu)Th1 CD4+ and CD8+ T cells in humanized mice. FINDINGS CD40.CoV2 vaccination selectively enriched long-lived spike- and nucleocapsid-specific CD8+ progenitors with stem-cell-like memory (Tscm) properties, whereas mRNA BNT162b2 induced effector memory CD8+ T cells. CD8+ Tscm cells produced IFNγ and TNF upon antigenic restimulation and showed a high proliferation rate. We demonstrate that CD40 activation is specifically required for the generation of huCD8+ Tscm cells. INTERPRETATION These results support the development of a CD40-vaccine platform capable of eliciting long-lasting T-cell immunity. FUNDING This work was supported by Inserm, Université Paris-Est Créteil, and the Investissements d'Avenir program, Vaccine Research Institute (VRI), managed by the ANR.
Collapse
Affiliation(s)
- Laury Nguema
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Florence Picard
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Marwa El Hajj
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Léa Dupaty
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Craig Fenwick
- Service of Immunology and Allergy Lausanne University Hospital, Swiss Vaccine Research Institute, University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Sylvain Cardinaud
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Aurélie Wiedemann
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Giuseppe Pantaleo
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; Service of Immunology and Allergy Lausanne University Hospital, Swiss Vaccine Research Institute, University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Sandra Zurawski
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; Baylor Scott and White Research Institute, Dallas, TX, United States
| | - Mireille Centlivre
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Gerard Zurawski
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; Baylor Scott and White Research Institute, Dallas, TX, United States
| | - Yves Lévy
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; Assistance Publique-Hôpitaux de Paris, Groupe Henri-Mondor Albert-Chenevier, Service Immunologie Clinique, Créteil, France.
| | - Véronique Godot
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France.
| |
Collapse
|
31
|
Mazzotti L, Borges de Souza P, Azzali I, Angeli D, Nanni O, Sambri V, Semprini S, Bravaccini S, Cerchione C, Gaimari A, Nicolini F, Ancarani V, Martinelli G, Pasetto A, Calderon H, Juan M, Mazza M. Exploring the Relationship Between Humoral and Cellular T Cell Responses Against SARS-CoV-2 in Exposed Individuals From Emilia Romagna Region and COVID-19 Severity. HLA 2025; 105:e70011. [PMID: 39807702 PMCID: PMC11731316 DOI: 10.1111/tan.70011] [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: 07/09/2024] [Revised: 11/03/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025]
Abstract
COVID-19 remains a significant global health problem with uncertain long-term consequences for convalescents. We investigated the relationships between anti-N protein antibody levels, severe acute respiratory syndrome (SARS)-CoV-2-associated TCR repertoire parameters, HLA type and epidemiological information from three cohorts of 524 SARS-CoV-2-infected subjects subgrouped in acute phase, seronegative and seropositive convalescents from the Emilia Romagna region. Epidemiological information and anti-N antibody index were associated with TCR repertoire data. HLA type was inferred from TCR repertoire using the HLA3 tool and its association with clonal breadth (CB) and clonal depth (CD) was assessed. Age above 58 years, male and COVID-19 hospitalisation were significantly and independently associated with seropositivity (p = 0.004; p = 0.004; p = 0.04), suggesting an association between high antibody titres and symptoms' severity. As for the TCR repertoire analysis, we found no difference in CB among the cohorts, while CD was higher in seronegative than acute (p = 0.04). However, clustering analysis supported that seronegative patients are endowed with broader CB and deeper CD indicating a compensatory mechanism without effective seroconversion. The CD calculated on the TCRs associated with the single SARS-CoV-2 ORFs in convalescents is higher when compared to the acute. Lastly, we identified and reported on novel HLAs significantly associated with increased risk of hospitalisation such as HLA-C*07:02 carriers (OR = 3.9, CI = 1.1-13.4, p = 0.03) and on HLAs that associate significantly with lower or higher TCR repertoire parameters in a population exposed for the first time to SARS-CoV-2.
Collapse
Affiliation(s)
- Lucia Mazzotti
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | | | - Irene Azzali
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Davide Angeli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Oriana Nanni
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Vittorio Sambri
- Microbiology UnitThe Great Romagna Area Hub LaboratoryPievesestinaItaly
- DIMECBologna UniversityBolognaItaly
| | - Simona Semprini
- Microbiology UnitThe Great Romagna Area Hub LaboratoryPievesestinaItaly
| | - Sara Bravaccini
- Department of Medicine and SurgeryUniversity of Enna “Kore”EnnaItaly
| | - Claudio Cerchione
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Anna Gaimari
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Fabio Nicolini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Valentina Ancarani
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Giovanni Martinelli
- Department of Hematology and Sciences OncologyInstitute of Haematology “L. and A. Seràgnoli” S. Orsola, University Hospital in BolognaBolognaItaly
| | - Anna Pasetto
- Section for Cell TherapyRadiumhospitalet, Oslo University HospitalOsloNorway
- Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | - Hugo Calderon
- Department of ImmunologyCentre de Diagnòstic Biomèdic, Hospital Clínic of BarcelonaBarcelonaSpain
| | - Manel Juan
- Department of ImmunologyCentre de Diagnòstic Biomèdic, Hospital Clínic of BarcelonaBarcelonaSpain
| | - Massimiliano Mazza
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| |
Collapse
|
32
|
Genova SN, Pencheva MM, Abadjieva TI, Atanasov NG. Cellular and immune response in fatal COVID-19 pneumonia. Pan Afr Med J 2024; 49:130. [PMID: 40190436 PMCID: PMC11971930 DOI: 10.11604/pamj.2024.49.130.45739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/03/2024] [Indexed: 04/09/2025] Open
Abstract
Introduction the severity of COVID-19, causing fatal pneumonia, acute respiratory distress syndrome (ARDS), and thrombotic complications, is linked to intense inflammation. Elevated CD4+ and CD8+ cells in the lungs indicate harmful inflammation in severe cases. This study investigates immune responses in lung tissues of deceased patients across different stages of COVID-19 pneumonia. Methods lung tissues from 160 fatal COVID-19 cases, diagnosed via Real-Time RT-PCR, were histologically analyzed to identify pneumonia stages. Inflammatory cell counts were assessed immunohistochemically. Non-parametric tests analyzed categorical variables, while regression analysis evaluated relationships between continuous variables. Results the average patient age was 68.1 years (± 12.6). Microscopic analysis identified four pneumonia stages. CD4+, CD68 (macrophages), and IgG4 levels peaked by day 14, with notable elevation within seven days of symptom onset. CD4+ levels were significantly lower in DAD pneumonia (49.4% ± 15.7%) compared to ARDS (66.4% ± 19.3%) and thrombosis (70.2% ± 28.9%) (p < 0.05). Male patients had higher CD4+ values (68.5% ± 21.1%) than females (56.9% ± 22.4%) (p < 0.05). B cells (CD20) and NK cells were depleted across all stages. IgG4 expression reached 80-90% in acute phases but was nearly absent during organization and fibrosis stages. Conclusion a sharp decline in CD4+ and CD8+ during acute pneumonia and sepsis reflects immune exhaustion, while their elevation in ARDS and thrombosis likely triggers cytokine storms, causing severe lung damage. Elevated IgG4 levels in acute lung tissue correlate with fatal outcomes in severe COVID-19.
Collapse
Affiliation(s)
- Sylvia Nikolaeva Genova
- Department of General and Clinical Pathology, Medical Faculty, Medical University Plovdiv, Plovdiv, Bulgaria
- St George University Hospital, Plovdiv, Bulgaria
| | - Mina Miroslavova Pencheva
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Tsvetana Ivanova Abadjieva
- St George University Hospital, Plovdiv, Bulgaria
- Department of Dermatology and Venereology, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Nikolay Georgiev Atanasov
- Department of Health Management and Health Economics, Faculty of Public Health, Medical University Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
33
|
Nakayama EE, Shioda T. Detrimental Effects of Anti-Nucleocapsid Antibodies in SARS-CoV-2 Infection, Reinfection, and the Post-Acute Sequelae of COVID-19. Pathogens 2024; 13:1109. [PMID: 39770368 PMCID: PMC11728538 DOI: 10.3390/pathogens13121109] [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: 11/12/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Antibody-dependent enhancement (ADE) is a phenomenon in which antibodies enhance subsequent viral infections rather than preventing them. Sub-optimal levels of neutralizing antibodies in individuals infected with dengue virus are known to be associated with severe disease upon reinfection with a different dengue virus serotype. For Severe Acute Respiratory Syndrome Coronavirus type-2 infection, three types of ADE have been proposed: (1) Fc receptor-dependent ADE of infection in cells expressing Fc receptors, such as macrophages by anti-spike antibodies, (2) Fc receptor-independent ADE of infection in epithelial cells by anti-spike antibodies, and (3) Fc receptor-dependent ADE of cytokine production in cells expressing Fc receptors, such as macrophages by anti-nucleocapsid antibodies. This review focuses on the Fc receptor-dependent ADE of cytokine production induced by anti-nucleocapsid antibodies, examining its potential role in severe COVID-19 during reinfection and its contribution to the post-acute sequelae of COVID-19, i.e., prolonged symptoms lasting at least three months after the acute phase of the disease. We also discuss the protective effects of recently identified anti-spike antibodies that neutralize Omicron variants.
Collapse
Affiliation(s)
| | - Tatsuo Shioda
- Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan;
| |
Collapse
|
34
|
Meyer NJ, Prescott HC. Sepsis and Septic Shock. N Engl J Med 2024; 391:2133-2146. [PMID: 39774315 DOI: 10.1056/nejmra2403213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Affiliation(s)
- Nuala J Meyer
- From the Division of Pulmonary, Allergy, and Critical Care Medicine and the Center for Translational Lung Biology, Lung Biology Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia (N.J.M.); and the Department of Internal Medicine, University of Michigan, and VA Center for Clinical Management Research - both in Ann Arbor (H.C.P.)
| | - Hallie C Prescott
- From the Division of Pulmonary, Allergy, and Critical Care Medicine and the Center for Translational Lung Biology, Lung Biology Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia (N.J.M.); and the Department of Internal Medicine, University of Michigan, and VA Center for Clinical Management Research - both in Ann Arbor (H.C.P.)
| |
Collapse
|
35
|
Hasan S, Awasthi P, Malik S, Dwivedi M. Immunotherapeutic strategies to induce inflection in the immune response: therapy for cancer and COVID-19. Biotechnol Genet Eng Rev 2024; 40:3571-3610. [PMID: 36411974 DOI: 10.1080/02648725.2022.2147661] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022]
Abstract
Cancer has agonized the human race for millions of years. The present decade witnesses biological therapeutics to combat cancer effectively. Cancer Immunotherapy involves the use of therapeutics for manipulation of the immune system by immune agents like cytokines, vaccines, and transfection agents. Recently, this therapeutic approach has got vast attention due to the current pandemic COVID-19 and has been very effective. Concerning cancer, immunotherapy is based on the activation of the host's antitumor response by enhancing effector cell number and the production of soluble mediators, thereby reducing the host's suppressor mechanisms by induction of a tumour killing environment and by modulating immune checkpoints. In the present era, immunotherapies have gained traction and momentum as a pedestal of cancer treatment, improving the prognosis of many patients with a wide variety of haematological and solid malignancies. Food supplements, natural immunomodulatory drugs, and phytochemicals, with recent developments, have shown positive trends in cancer treatment by improving the immune system. The current review presents the systematic studies on major immunotherapeutics and their development for the effective treatment of cancers as well as in COVID-19. The focus of the review is to highlight comparative analytics of existing and novel immunotherapies in cancers, concerning immunomodulatory drugs and natural immunosuppressants, including immunotherapy in COVID-19 patients.
Collapse
Affiliation(s)
- Saba Hasan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Prankur Awasthi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University, Ranchi, Jharkhand, India
| | - Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| |
Collapse
|
36
|
Sabino‐Santos G, Leggio CE, Litwin SM, Waheed N, Bai S, Ulusan S, Karunathilake A, Elliott DH, Smira AR, Chandra S, Li L, Ning B, Hu T, Schieffelin JS, Gunn BM, Robinson JE, Fuloria J, Norton EB. Post-COVID immunity in patients with solid tumor or hematological malignancies treated with SARS-CoV-2 monoclonal antibodies. Immun Inflamm Dis 2024; 12:e70039. [PMID: 39659018 PMCID: PMC11632117 DOI: 10.1002/iid3.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/12/2024] [Accepted: 10/01/2024] [Indexed: 12/12/2024] Open
Abstract
PURPOSE SARS-CoV-2 monoclonal antibody (mAB) therapy has effectively treated severe COVID-19, although how this contributes to protective antiviral immunity in settings of malignancy is poorly defined. PATIENTS AND METHODS We evaluated the development of post-infection immunity in five patients with malignancies who received mAB therapy targeting spike protein for their PCR-confirmed SARS-CoV-2 infection in 2021, compared with non-mAB controls. Patients were identified from a larger study on oncology with a history or documented current infection with SARS-CoV-2. Subjects include two patients with lymphoma and CD20-depletion therapy, one with myeloma and two with solid tumor (stage IIA rectal adenocarcinoma and metastatic breast cancer). Cancer therapies and COVID vaccination history varied by patient. Blood samples (1-4 per patient) were collected 71-635 days post-mAB therapy. We employed clinical histories with comprehensive immunoprofiling analysis, including systems serology antibody isotyping and effector function, T-cell immunophenotyping for subset and memory cells, and sensitive blood viral RNA detection up to 2 years post-mAB therapy. RESULTS B-cell deficiency was confirmed in 3/5 patients. All patients had detectable anti-spike and nucleoprotein antibody isotypes, effector functions, and neutralizing antibodies (which increased over time by subject) at similar levels to the control group. Virus-specific T-cell activation and phenotypes varied by time and patient. Spike-specific effector and memory CD8 + T-cells were significantly elevated in mAB subjects compared to the control group. SARS-CoV-2 viral RNA detection was also higher in mAB-treated patients. One patient on bortezomib therapy had unique alterations in these populations. CONCLUSION All mAB-treated patients with malignancies developed polyfunctional immunity humoral and T-cell immunity to SARS-CoV-2 even in the setting of B-cell deficiency. The evolution of this immunity, including new variant-specific antibodies, without secondary illnesses suggests that patients were protected from symptomatic re-infection, and mAB therapy did not blunt the development of host immunity. Future studies are warranted to better characterize immunologic memory over time with exposures to new viral variants, evaluate prolonged viral shedding and the continued use of appropriate mAB for infection in high-risk patients.
Collapse
Affiliation(s)
- Gilberto Sabino‐Santos
- Department of Microbiology and ImmunologyTulane University School of MedicineNew OrleansLouisianaUSA
| | | | - Sean M. Litwin
- Department of Microbiology and ImmunologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Najia Waheed
- University Medical Center New OrleansNew OrleansLouisianaUSA
| | - Shuangyi Bai
- Paul G. Allen School of Global HealthWashington State UniversityPullmanWashingtonUSA
| | - Sinem Ulusan
- Paul G. Allen School of Global HealthWashington State UniversityPullmanWashingtonUSA
| | - Anoli Karunathilake
- Paul G. Allen School of Global HealthWashington State UniversityPullmanWashingtonUSA
| | - Debra H. Elliott
- Department of PediatricsTulane University School of MedicineNew OrleansLouisianaUSA
| | - Ashley R. Smira
- Department of PediatricsTulane University School of MedicineNew OrleansLouisianaUSA
| | - Sruti Chandra
- Department of Microbiology and ImmunologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Lin Li
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Bo Ning
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Tony Hu
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - John S. Schieffelin
- Department of PediatricsTulane University School of MedicineNew OrleansLouisianaUSA
| | - Bronwyn M. Gunn
- Paul G. Allen School of Global HealthWashington State UniversityPullmanWashingtonUSA
| | - James E. Robinson
- Department of PediatricsTulane University School of MedicineNew OrleansLouisianaUSA
| | - Jyotsna Fuloria
- University Medical Center New OrleansNew OrleansLouisianaUSA
| | - Elizabeth B. Norton
- Department of Microbiology and ImmunologyTulane University School of MedicineNew OrleansLouisianaUSA
| |
Collapse
|
37
|
Amarilla-Irusta A, Zenarruzabeitia O, Sevilla A, Sandá V, Lopez-Pardo A, Astarloa-Pando G, Pérez-Garay R, Pérez-Fernández S, Meijide S, Imaz-Ayo N, Arana-Arri E, Amo L, Borrego F. CD151 identifies an NK cell subset that is enriched in COVID-19 patients and correlates with disease severity. J Infect 2024; 89:106304. [PMID: 39374860 DOI: 10.1016/j.jinf.2024.106304] [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: 04/22/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/09/2024]
Abstract
Severe coronavirus disease 2019 (COVID-19) often leads to acute respiratory distress syndrome and multi-organ dysfunction, driven by a dysregulated immune response, including a cytokine storm with elevated proinflammatory cytokine levels. Natural killer (NK) cells are part of the innate immune system with a fundamental role in the defense against viral infections. However, during COVID-19 acute infection, they exhibit an altered phenotype and impaired functionality contributing to the immunopathogenesis of the disease. In this work, we have studied a cohort of patients with COVID-19 (ranging from mild to severe) by analyzing IL-15, TGF-β, PlGF and GDF-15 plasma levels and performing multiparametric flow cytometry studies. Our results revealed that severe COVID-19 patients exhibited high levels of IL-15, PlGF and GDF-15, along with an enrichment of an NK cell subset expressing the CD151 tetraspanin, which correlated with IL-15 plasma levels and disease severity. In patients, these CD151+ NK cells displayed a more activated phenotype characterized by an increased expression of HLA-DR, CD38 and granzyme B, a distinct receptor repertoire, with lower levels of CD160 and CD31 and higher levels of CD55 and, remarkably, a higher expression of tissue-resident markers CD103 and the NK cell decidual marker CD9. Last of all, in individuals with severe disease, we identified an expansion of a CD151brightCD9+ NK cell subset, suggesting that these cells play a specific role in COVID-19. Altogether, our findings suggest that CD151+ NK cells may have a relevant role in COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Arrate Sevilla
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Víctor Sandá
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Ainara Lopez-Pardo
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | | | - Raquel Pérez-Garay
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Clinical Analysis Service, Cruces University Hospital, OSI Ezkerraldea-Enkarterri-Cruces, Barakaldo, Spain
| | - Silvia Pérez-Fernández
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Susana Meijide
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Natale Imaz-Ayo
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Eunate Arana-Arri
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Laura Amo
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Francisco Borrego
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
38
|
Dave RH, Crayton H, Miravalle A, Tai MH, Wyse K, Houghton K, Hitchens A, Berkovich R. COVID-19 Vaccination Response in Patients with Multiple Sclerosis Treated with Ofatumumab in the United States: A Medical Record Review. Neurol Ther 2024; 13:1737-1745. [PMID: 39441497 PMCID: PMC11541983 DOI: 10.1007/s40120-024-00671-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION Real-world data are required to provide a greater understanding of the impact of ofatumumab on the ability to mount an effective immune response following the receipt of approved COVID-19 vaccinations. This retrospective real-world analysis aimed to describe the humoral immune response to COVID-19 vaccination during ofatumumab treatment in patients with multiple sclerosis (MS). METHODS Data from patients with MS treated with ofatumumab who were fully vaccinated against COVID-19 infection were abstracted from medical charts at four clinical sites in the USA. Patient characteristics and humoral response were summarized descriptively. Differences in humoral response were documented on the basis of vaccination status during ofatumumab treatment (i.e., after full vaccination and after booster vaccination) and prior disease-modifying treatment (DMT) exposure (i.e., DMT naïve, prior anti-CD20/sphingosine 1-phosphate [S1P] therapy, prior non-anti-CD20/S1P therapy). The sample size precluded formal statistical analysis. RESULTS Thirty-eight patients were included. The mean (standard deviation) duration of ofatumumab treatment upon data collection was 20.4 (4.6) months (treatment ongoing for 35 [92%] patients). Definitive humoral response after full vaccination was documented for 34 patients, of whom 20 (60%) were seropositive. Definitive humoral response after booster vaccination was documented among five patients, of whom three (60%) were seropositive. Among patients who were DMT naïve prior to ofatumumab (n = 15), 73% were seropositive; among patients exposed to prior anti-CD20/S1P therapy (n = 14), 33% were seropositive; and among patients exposed to prior non-anti-CD20/S1P therapy (n = 9), 56% were seropositive. Patients naïve to DMT had been living with an MS diagnosis for a shorter duration than those experienced with DMTs. CONCLUSION Patients with MS receiving ongoing treatment with ofatumumab can mount a positive humoral response to a COVID-19 vaccination. Prior treatment with anti-CD20 or S1P DMTs may be a risk factor for lower humoral response.
Collapse
Affiliation(s)
- Rahul H Dave
- Inova Medical Group-Neurology II, Fairfax, VA, USA
| | - Heidi Crayton
- Multiple Sclerosis Center of Greater Washington, Washington, VA, USA
| | | | - Ming-Hui Tai
- Novartis Pharmaceuticals Corporation, One Health Plaza East, Hanover, NJ, 07936, USA.
| | - Kerri Wyse
- Novartis Pharmaceuticals Corporation, One Health Plaza East, Hanover, NJ, 07936, USA
| | | | | | - Regina Berkovich
- Los Angeles County General Hospital and Clinic, Los Angeles, CA, USA
| |
Collapse
|
39
|
Li Y, Chen Y, Liang J, Wang Y. Immunological characteristics in elderly COVID-19 patients: a post-COVID era analysis. Front Cell Infect Microbiol 2024; 14:1450196. [PMID: 39679195 PMCID: PMC11638707 DOI: 10.3389/fcimb.2024.1450196] [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: 06/17/2024] [Accepted: 10/14/2024] [Indexed: 12/17/2024] Open
Abstract
Background Advanced age is a primary risk factor for adverse COVID-19 outcomes, potentially attributed to immunosenescence and dysregulated inflammatory responses. In the post-pandemic era, with containment measures lifted, the elderly remain particularly susceptible, highlighting the need for intensified focus on immune health management. Methods A total of 281 elderly patients were enrolled in this study and categorized based on their clinical status at the time of admission into three groups: non-severe (n = 212), severe survivors (n = 49), and severe non-survivors (n = 20). Binary logistic regression analysis was employed to identify independent risk factors associated with disease severity and in-hospital outcomes. The diagnostic performance of risk factors was assessed using the receiver operating characteristic (ROC) curves. Kaplan-Meier survival analysis and log-rank test were utilized to compare the 30-day survival rates. Furthermore, the transcriptomic data of CD4+ T cells were extracted from Gene Expression Omnibus (GEO) database. Gene Set Enrichment Analysis (GSEA) was applied to reveal biological processes and pathways involved. Results In the comparison between severe and non-severe COVID-19 cases, significant elevations were observed in the neutrophil-to-lymphocyte ratio (NLR), C-reactive protein (CRP), and Serum Amyloid A (SAA) levels, concurrent with a notable reduction in CD8+ T cells, CD4+ T cells, natural killer (NK) cells, and monocytes (all p < 0.05). CD4+ T cells (OR: 0.997 [0.995-1.000], p<0.05) and NLR (OR: 1.03 [1.001-1.060], p<0.05) were independent risk factors affecting disease severity. The diagnostic accuracy for COVID-19 severity, as measured by the area under the curve (AUC) for CD4+ T cells and NLR, was 0.715 (95% CI: 0.645-0.784) and 0.741 (95% CI: 0.675-0.807), respectively. Moreover, patients with elevated NLR or IL-6 levels at admission exhibited significantly shorter survival times. Gene Set Enrichment Analysis (GSEA) revealed several biological pathways that are implicated in the regulation of immune responses and metabolic processes. Conclusions Lymphocytopenia and the cytokine storm onset are significant predictors of an unfavorable prognosis in elderly patients. The decrease in CD4+ T cells among elderly patients is detrimental to disease recovery, and the biological pathways regulated by these cells could potentially heighten vulnerability to SARS-CoV-2 infection, thereby exacerbating the development of associated complications.
Collapse
Affiliation(s)
| | | | | | - Yajie Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Strumiliene E, Urbonienė J, Jurgauskiene L, Zeleckiene I, Bliudzius R, Malinauskiene L, Zablockiene B, Samuilis A, Jancoriene L. Long-Term Pulmonary Sequelae and Immunological Markers in Patients Recovering from Severe and Critical COVID-19 Pneumonia: A Comprehensive Follow-Up Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1954. [PMID: 39768836 PMCID: PMC11678914 DOI: 10.3390/medicina60121954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: Severe and critical COVID-19 pneumonia can lead to long-term complications, especially affecting pulmonary function and immune health. However, the extent and progression of these complications over time are not well understood. This study aimed to assess lung function, radiological changes, and some immune parameters in survivors of severe and critical COVID-19 up to 12 months after hospital discharge. Materials and Methods: This prospective observational cohort study followed 85 adult patients who were hospitalized with severe or critical COVID-19 pneumonia at a tertiary care hospital in Vilnius, Lithuania, for 12 months post-discharge. Pulmonary function tests (PFTs), including forced vital capacity (FVC), forced expiratory volume in 1 s (FEV1), and diffusion capacity for carbon monoxide (DLCO), were conducted at 3, 6, and 12 months. High-resolution chest computed tomography (CT) scans assessed residual inflammatory and profibrotic/fibrotic abnormalities. Lymphocyte subpopulations were evaluated via flow cytometry during follow-up visits to monitor immune status. Results: The median age of the cohort was 59 years (IQR: 51-64). Fifty-three (62.4%) patients had critical COVID-19 disease. Pulmonary function improved significantly over time, with increases in FVC, FEV1, VC, TLC, and DLCO. Residual volume (RV) did not change significantly over time, suggesting that some aspects of lung function, such as air trapping, remained stable and may require attention in follow-up care. The percentage of patients with restrictive spirometry patterns decreased from 24.71% at 3 months to 14.8% at 12 months (p < 0.05). Residual inflammatory changes on CT were present in 77.63% at 6 months, decreasing to 69.62% at 12 months (p < 0.001). Profibrotic changes remained prevalent, affecting 82.89% of patients at 6 months and 73.08% at 12 months. Lymphocyte counts declined significantly from 3 to 12 months (2077 cells/µL vs. 1845 cells/µL, p = 0.034), with notable reductions in CD3+ (p = 0.040), CD8+ (p = 0.007), and activated CD3HLA-DR+ cells (p < 0.001). This study found that higher CD4+ T cell counts were associated with worse lung function, particularly reduced total lung capacity (TLC), while higher CD8+ T cell levels were linked to improved pulmonary outcomes, such as increased forced vital capacity (FVC) and vital capacity (VC). Multivariable regression analyses revealed that increased levels of CD4+/CD28+/CD192+ T cells were associated with worsening lung function, while higher CD8+/CD28+/CD192+ T cell counts were linked to better pulmonary outcomes, indicating that immune dysregulation plays a critical role in long-term respiratory recovery. Conclusions: Survivors of severe and critical COVID-19 pneumonia continue to experience significant long-term impairments in lung function and immune system health. Regular monitoring of pulmonary function, radiological changes, and immune parameters is essential for guiding personalized post-COVID-19 care and improving long-term outcomes. Further research is needed to explore the mechanisms behind these complications and to develop targeted interventions for long COVID-19.
Collapse
Affiliation(s)
- Edita Strumiliene
- Clinic of Infectious Diseases and Dermatovenerology, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, 01513 Vilnius, Lithuania;
| | - Jurgita Urbonienė
- Centre of Infectious Diseases, Vilnius University Hospital Santaros klinikos, 08661 Vilnius, Lithuania;
| | - Laimute Jurgauskiene
- Clinic of Cardiac and Vascular Diseases, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, 01513 Vilnius, Lithuania;
| | - Ingrida Zeleckiene
- Department of Radiology, Nuclear Medicine and Medical Physics, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, 01513 Vilnius, Lithuania; (I.Z.); (R.B.); (A.S.)
| | - Rytis Bliudzius
- Department of Radiology, Nuclear Medicine and Medical Physics, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, 01513 Vilnius, Lithuania; (I.Z.); (R.B.); (A.S.)
| | - Laura Malinauskiene
- Clinic of Chest Diseases, Immunology and Allergology, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, 01513 Vilnius, Lithuania;
| | - Birutė Zablockiene
- Clinic of Infectious Diseases and Dermatovenerology, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, 01513 Vilnius, Lithuania;
| | - Arturas Samuilis
- Department of Radiology, Nuclear Medicine and Medical Physics, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, 01513 Vilnius, Lithuania; (I.Z.); (R.B.); (A.S.)
| | - Ligita Jancoriene
- Clinic of Infectious Diseases and Dermatovenerology, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, 01513 Vilnius, Lithuania;
| |
Collapse
|
41
|
Oh DS, Kim E, Normand R, Lu G, Shook LL, Lyall A, Jasset O, Demidkin S, Gilbert E, Kim J, Akinwunmi B, Tantivit J, Tirard A, Arnold BY, Slowikowski K, Goldberg MB, Filbin MR, Hacohen N, Nguyen LH, Chan AT, Yu XG, Li JZ, Yonker L, Fasano A, Perlis RH, Pasternak O, Gray KJ, Choi GB, Drew DA, Sen P, Villani AC, Edlow AG, Huh JR. SARS-CoV-2 infection elucidates features of pregnancy-specific immunity. Cell Rep 2024; 43:114933. [PMID: 39504241 PMCID: PMC11724703 DOI: 10.1016/j.celrep.2024.114933] [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: 12/14/2023] [Revised: 07/30/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
Pregnancy is a risk factor for increased severity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other respiratory infections, but the mechanisms underlying this risk are poorly understood. To gain insight into the role of pregnancy in modulating immune responses at baseline and upon SARS-CoV-2 infection, we collected peripheral blood mononuclear cells and plasma from 226 women, including 152 pregnant individuals and 74 non-pregnant women. We find that SARS-CoV-2 infection is associated with altered T cell responses in pregnant women, including a clonal expansion of CD4-expressing CD8+ T cells, diminished interferon responses, and profound suppression of monocyte function. We also identify shifts in cytokine and chemokine levels in the sera of pregnant individuals, including a robust increase of interleukin-27, known to drive T cell exhaustion. Our findings reveal nuanced pregnancy-associated immune responses, which may contribute to the increased susceptibility of pregnant individuals to viral respiratory infection.
Collapse
Affiliation(s)
- Dong Sun Oh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Eunha Kim
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; BK21 Graduate Program, Department of Biomedical Sciences and Department of Neuroscience, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Rachelly Normand
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Guangqing Lu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Lydia L Shook
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amanda Lyall
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Olyvia Jasset
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stepan Demidkin
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Emily Gilbert
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Joon Kim
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Babatunde Akinwunmi
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jessica Tantivit
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alice Tirard
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Benjamin Y Arnold
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kamil Slowikowski
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Marcia B Goldberg
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA; Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Michael R Filbin
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nir Hacohen
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Long H Nguyen
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Xu G Yu
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Jonathan Z Li
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lael Yonker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Roy H Perlis
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kathryn J Gray
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Gloria B Choi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David A Drew
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Pritha Sen
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA; Transplant, Oncology, and Immunocompromised Host Group, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Alexandra-Chloé Villani
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - Andrea G Edlow
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
42
|
An H, Li T, Zhang X, Hu H, Zhang C, Wang Y, Jin S, Li M. Persistent CD19 + B cell lymphopenia in critically ill COVID-19 patients 50 days after symptom onset. Front Cell Infect Microbiol 2024; 14:1488607. [PMID: 39650838 PMCID: PMC11621214 DOI: 10.3389/fcimb.2024.1488607] [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: 08/30/2024] [Accepted: 11/08/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction Long COVID (LC) poses a persistent challenge in clinical practice due to limited understanding of its etiology. LC is hypothesized to stem from aberrant immune responses in COVID-19. Vaccinations, which boost immune cells to restore function, could help ease LC symptoms. Methods To exclude the impact of vaccination, we examined the immune cell profiles of recovering COVID-19 patients before vaccines were available. White blood cell differentials were monitored in ninety-twohealthy unvaccinated controls. Seventy-six unvaccinated COVID-19 patients were monitored upon admission and on the 50th day post-symptom onset (DPSO50). Peripheral lymphocyte subsets were analyzed using flow cytometry. Results Mild cases showed no significant changes in lymphocyte counts or subsets from admission to DPSO50. By DPSO50, severe and critical cases showed almost complete recovery from lymphopenia, with critical cases having CD19+ B-cell counts approximately 45% lower than the mild group. Severe and critical cases exhibited reduced B-cell frequencies, with critical cases displaying around 48% higher natural killer (NK) cell counts. In mild cases, NK cell counts negatively correlated with B-cell counts (r=-0.528, p=0.02). Additionally, critical cases showed positive correlations between NK cell counts and CD4+ T-cell counts (r=0.83, p<0.01), and between NK cell counts and CD8+ T-cell counts (r=0.74, p<0.01). Severe cases demonstrated decreased counts of CD4+CD25+CD127lowFoxP3+ regulatory T-cells (Tregs), which positively correlated with B-cell counts (r=0.37, p<0.05). Discussion Our findings indicate that aberrant immune cell profiles in COVID-19 patients change dynamically during recovery, depending on disease severity. This study suggests that convalescent patients from critical COVID-19 may experience long-lasting B-cell lymphopenia.
Collapse
Affiliation(s)
- Hui An
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ting Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinyue Zhang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hao Hu
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chen Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongyu Wang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shengwei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ming Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
43
|
Kim J, Ionita M, Lee M, McKeague ML, Pattekar A, Painter MM, Wagenaar J, Truong V, Norton DT, Mathew D, Nam Y, Apostolidis SA, Clendenin C, Orzechowski P, Jung SH, Woerner J, Ittner CAG, Turner AP, Esperanza M, Dunn TG, Mangalmurti NS, Reilly JP, Meyer NJ, Calfee CS, Liu KD, Matthy MA, Swigart LB, Burnham EL, McKeehan J, Gandotra S, Russel DW, Gibbs KW, Thomas KW, Barot H, Greenplate AR, Wherry EJ, Kim D. Cytometry masked autoencoder: An accurate and interpretable automated immunophenotyper. Cell Rep Med 2024; 5:101808. [PMID: 39515318 PMCID: PMC11604491 DOI: 10.1016/j.xcrm.2024.101808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/09/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Single-cell cytometry data are crucial for understanding the role of the immune system in diseases and responses to treatment. However, traditional methods for annotating cytometry data face challenges in scalability, robustness, and accuracy. We propose a cytometry masked autoencoder (cyMAE), which automates immunophenotyping tasks including cell type annotation. The model upholds user-defined cell type definitions, facilitating interpretability and cross-study comparisons. The training of cyMAE has a self-supervised phase, which leverages large amounts of unlabeled data, followed by fine-tuning on specialized tasks using smaller amounts of annotated data. The cost of training a new model is amortized over repeated inferences on new datasets using the same panel. Through validation across multiple studies using the same panel, we demonstrate that cyMAE delivers accurate and interpretable cellular immunophenotyping and improves the prediction of subject-level metadata. This proof of concept marks a significant step forward for large-scale immunology studies.
Collapse
Affiliation(s)
- Jaesik Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matei Ionita
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew Lee
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michelle L McKeague
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ajinkya Pattekar
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark M Painter
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joost Wagenaar
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Van Truong
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dylan T Norton
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Divij Mathew
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yonghyun Nam
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sokratis A Apostolidis
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Rheumatology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cynthia Clendenin
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patryk Orzechowski
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Automatics and Robotics, AGH University of Science and Technology, al. Mickiewicza 30, 30-059 Kraków, Poland
| | - Sang-Hyuk Jung
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jakob Woerner
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Caroline A G Ittner
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandra P Turner
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mika Esperanza
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas G Dunn
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nilam S Mangalmurti
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John P Reilly
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nuala J Meyer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Carolyn S Calfee
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, School of Medicine, San Francisco, CA 94143, USA; Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, University of California, San Francisco, School of Medicine, San Francisco, CA 94143, USA; Cardiovascular Research Institute, Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, CA 94158, USA
| | - Kathleen D Liu
- Division of Nephrology and Critical Care Medicine, University of California, San Francisco, School of Medicine, San Francisco, CA 94143, USA
| | - Michael A Matthy
- Cardiovascular Research Institute, Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, CA 94158, USA
| | - Lamorna Brown Swigart
- Department of Laboratory Medicine, University of California, San Francisco, School of Medicine, San Francisco, CA 94143, USA
| | - Ellen L Burnham
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jeffrey McKeehan
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Sheetal Gandotra
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Derek W Russel
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Pulmonary Section, Birmingham Veteran's Affairs Medical Center, Birmingham, AL 35233, USA
| | - Kevin W Gibbs
- Section on Pulmonary and Critical Care, Allergy, and Immunology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Karl W Thomas
- Section on Pulmonary and Critical Care, Allergy, and Immunology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Harsh Barot
- Section on Hospital Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Allison R Greenplate
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - E John Wherry
- Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Dokyoon Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology & Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
44
|
de Oliveira G, Costa-Rocha IA, Oliveira-Carvalho N, dos Santos TMAF, Campi-Azevedo AC, Peruhype-Magalhães V, Miranda VHS, Prado RO, Pereira AAS, Alves CC, Brito-de-Sousa JP, Reis LR, Costa-Pereira C, da Mata CPSM, Almeida VES, dos Santos LM, Almeida GG, Antonelli LRDV, Coelho-dos-Reis JG, Teixeira-Carvalho A, Martins-Filho OA. Phenotypic Timeline Kinetics, Integrative Networks, and Performance of T- and B-Cell Subsets Associated with Distinct Clinical Outcome of Severe COVID-19 Patients. Microorganisms 2024; 12:2272. [PMID: 39597661 PMCID: PMC11596994 DOI: 10.3390/microorganisms12112272] [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: 05/29/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 11/29/2024] Open
Abstract
The present study aimed to evaluate the kinetics of the phenotypic profile and integrative networks of T/B-cells in severe COVID-19 patients, categorized according to disease outcome, during the circulation of the B.1.1.28 and B.1.1.33 SARS-CoV-2 strains in Brazil. Peripheral blood obtained at distinct time points (baseline/D0; D7; D14-28) was used for ex vivo flow cytometry immunophenotyping. The data demonstrated a decrease at D0 in the frequency of CD3+ T-cells and CD4+ T-cells and an increase in B-cells with mixed activation/exhaustion profiles. Higher changes in B-cell and CD4+ T-cells at D7 were associated with discharge/death outcomes, respectively. Regardless of the lower T/B-cell connectivity at D0, distinct profiles from D7/D14-28 revealed that, while discharge was associated with increasing connectivity for B-cells, CD4+ and CD8+ T-cells death was related to increased connectivity involving B-cells, but with lower connections mediated by CD4+ T-cells. The CD4+CD38+ and CD8+CD69+ subsets accurately classified COVID-19 vs. healthy controls throughout the kinetic analysis. Binary logistic regression identified CD4+CD107a+, CD4+T-bet+, CD8+CD69+, and CD8+T-bet+ at D0 and CD4+CD45RO+CD27+ at D7 as subsets associated with disease outcomes. Results showed that distinct phenotypic timeline kinetics and integrative networks of T/B-cells are associated with COVID-19 outcomes that may subsidize the establishment of applicable biomarkers for clinical/therapeutic monitoring.
Collapse
Affiliation(s)
- Gabriela de Oliveira
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Ismael Artur Costa-Rocha
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Nani Oliveira-Carvalho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Tâmilla Mayane Alves Fidelis dos Santos
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Ana Carolina Campi-Azevedo
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Vanessa Peruhype-Magalhães
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Vitor Hugo Simões Miranda
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Roberta Oliveira Prado
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Agnes Antônia Sampaio Pereira
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Clarice Carvalho Alves
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Joaquim Pedro Brito-de-Sousa
- Departamento de Imunologia e Parasitologia, Universidade Federal de Uberlândia, Uberlândia 38408-100, MG, Brazil;
| | - Laise Rodrigues Reis
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Christiane Costa-Pereira
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | | | | | - Liliane Martins dos Santos
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Gregório Guilherme Almeida
- Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.G.A.); (L.R.d.V.A.)
| | - Lis Ribeiro do Valle Antonelli
- Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.G.A.); (L.R.d.V.A.)
| | - Jordana Grazziela Coelho-dos-Reis
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Andréa Teixeira-Carvalho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| |
Collapse
|
45
|
Zhao C, Bai Y, Wang W, Amonkar GM, Mou H, Olejnik J, Hume AJ, Mühlberger E, Lukacs NW, Fearns R, Lerou PH, Ai X. Activation of STAT3-mediated ciliated cell survival protects against severe infection by respiratory syncytial virus. J Clin Invest 2024; 134:e183978. [PMID: 39484716 PMCID: PMC11527452 DOI: 10.1172/jci183978] [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: 06/17/2024] [Accepted: 08/15/2024] [Indexed: 11/03/2024] Open
Abstract
Respiratory syncytial virus (RSV) selectively targets ciliated cells in human bronchial epithelium and can cause bronchiolitis and pneumonia, mostly in infants. To identify molecular targets of intervention during RSV infection in infants, we investigated how age regulates RSV interaction with the bronchial epithelium barrier. Employing precision-cut lung slices and air-liquid interface cultures generated from infant and adult human donors, we found robust RSV virus spread and extensive apoptotic cell death only in infant bronchial epithelium. In contrast, adult bronchial epithelium showed no barrier damage and limited RSV infection. Single nuclear RNA-Seq revealed age-related insufficiency of an antiapoptotic STAT3 activation response to RSV infection in infant ciliated cells, which was exploited to facilitate virus spread via the extruded apoptotic ciliated cells carrying RSV. Activation of STAT3 and blockade of apoptosis rendered protection against severe RSV infection in infant bronchial epithelium. Lastly, apoptotic inhibitor treatment of a neonatal mouse model of RSV infection mitigated infection and inflammation in the lung. Taken together, our findings identify a STAT3-mediated antiapoptosis pathway as a target to battle severe RSV disease in infants.
Collapse
Affiliation(s)
- Caiqi Zhao
- Division of Newborn Medicine, Department of Pediatrics and
| | - Yan Bai
- Division of Newborn Medicine, Department of Pediatrics and
| | - Wei Wang
- Division of Newborn Medicine, Department of Pediatrics and
| | | | - Hongmei Mou
- The Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, Massachusetts, USA
| | - Judith Olejnik
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Adam J. Hume
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Elke Mühlberger
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Nicholas W. Lukacs
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Rachel Fearns
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Paul H. Lerou
- Division of Newborn Medicine, Department of Pediatrics and
| | - Xingbin Ai
- Division of Newborn Medicine, Department of Pediatrics and
| |
Collapse
|
46
|
Günter M, Mueller KAL, Salazar MJ, Gekeler S, Prang C, Harm T, Gawaz MP, Autenrieth SE. Immune signature of patients with cardiovascular disease predicts increased risk for a severe course of COVID-19. Eur J Immunol 2024; 54:e2451145. [PMID: 39094122 DOI: 10.1002/eji.202451145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024]
Abstract
Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection can lead to life-threatening clinical manifestations. Patients with cardiovascular disease (CVD) are at higher risk for severe courses of COVID-19. So far, however, there are hardly any strategies for predicting the course of SARS-CoV-2 infection in CVD patients at hospital admission. Thus, we investigated whether this prediction is achievable by prospectively analysing the blood immunophenotype of 94 nonvaccinated participants, including uninfected and acutely SARS-CoV-2-infected CVD patients and healthy donors, using a 36-colour spectral flow cytometry panel. Unsupervised data analysis revealed little differences between healthy donors and CVD patients, whereas the distribution of the cell populations changed dramatically in SARS-CoV-2-infected CVD patients. The latter had more mature NK cells, activated monocyte subsets, central memory CD4+ T cells, and plasmablasts but fewer dendritic cells, CD16+ monocytes, innate lymphoid cells, and CD8+ T-cell subsets. Moreover, we identified an immune signature characterised by CD161+ T cells, intermediate effector CD8+ T cells, and natural killer T (NKT) cells that is predictive for CVD patients with a severe course of COVID-19. Thus, intensified immunophenotype analyses can help identify patients at risk of severe COVID-19 at hospital admission, improving clinical outcomes through specific treatment.
Collapse
Affiliation(s)
- Manina Günter
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
- German Cancer Research Centre, Research Group Dendritic Cells in Infection and Cancer, Heidelberg, Germany
| | - Karin Anne Lydia Mueller
- Department of Cardiology and Angiology, University Hospital Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Mathew J Salazar
- German Cancer Research Centre, Research Group Dendritic Cells in Infection and Cancer, Heidelberg, Germany
| | - Sarah Gekeler
- Department of Cardiology and Angiology, University Hospital Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Carolin Prang
- Department of Cardiology and Angiology, University Hospital Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Meinrad Paul Gawaz
- Department of Cardiology and Angiology, University Hospital Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Stella E Autenrieth
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
- German Cancer Research Centre, Research Group Dendritic Cells in Infection and Cancer, Heidelberg, Germany
| |
Collapse
|
47
|
Aquino A, Zaikova E, Kalinina O, Karonova TL, Rubinstein A, Mikhaylova AA, Kudryavtsev I, Golovkin AS. T Regulatory Cell Subsets Do Not Restore for One Year After Acute COVID-19. Int J Mol Sci 2024; 25:11759. [PMID: 39519310 PMCID: PMC11545974 DOI: 10.3390/ijms252111759] [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: 09/10/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
COVID-19, caused by SARS-CoV-2, triggers a complex immune response, with T regulatory cells (Tregs) playing a crucial role in maintaining immune homeostasis and preventing excessive inflammation. The current study investigates the function of T regulatory cells during COVID-19 infection and the subsequent recovery period, emphasizing their impact on immune regulation and inflammation control. We conducted a comprehensive analysis of Treg subpopulations in peripheral blood samples from COVID-19 patients at different stages: acute infection, early convalescence, and long-term recovery. Flow cytometry was employed to quantify Tregs including "naïve", central memory (CM), effector memory (EM), and terminally differentiated CD45RA+ effector cells (TEMRA). Additionally, the functional state of the Tregs was assessed by the expression of purinergic signaling molecules (CD39, CD73). Cytokine profiles were assessed through multiplex analysis. Our findings indicate a significant decrease in the number of Tregs during the acute phase of COVID-19, which correlates with heightened inflammatory markers and increased disease severity. Specifically, we found a decrease in the relative numbers of "naïve" and an increase in EM Tregs, as well as a decrease in the absolute numbers of "naïve" and CM Tregs. During the early convalescent period, the absolute counts of all Treg populations tended to increase, accompanied by a reduction in pro-inflammatory cytokines. Despite this, one year after recovery, the decreased subpopulations of regulatory T cells had not yet reached the levels observed in healthy donors. Finally, we observed the re-establishment of CD39 expression in all Treg subsets; however, there was no change in CD73 expression among Tregs. Understanding these immunological changes across different T regulatory subsets and adenosine signaling pathways offers important insights into the disease's pathogenesis and provides a broader view of immune system dynamics during recovery.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Alexey S. Golovkin
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (A.R.); (I.K.)
| |
Collapse
|
48
|
Vinutha M, Sharma UR, Swamy G, Rohini S, Vada S, Janandri S, Haribabu T, Taj N, Gayathri SV, Jyotsna SK, Mudagal MP. COVID-19-related liver injury: Mechanisms, diagnosis, management; its impact on pre-existing conditions, cancer and liver transplant: A comprehensive review. Life Sci 2024; 356:123022. [PMID: 39214285 DOI: 10.1016/j.lfs.2024.123022] [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: 02/19/2024] [Revised: 08/20/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
AIMS This review explores the mechanisms, diagnostic approaches, and management strategies for COVID-19-induced liver injury, with a focus on its impact on patients with pre-existing liver conditions, liver cancer, and those undergoing liver transplantation. MATERIALS AND METHODS A comprehensive literature review included studies on clinical manifestations of liver injury due to COVID-19. Key areas examined were direct viral effects, drug-induced liver injury, cytokine storms, and impacts on individuals with chronic liver diseases, liver transplants, and the role of vaccination. Data were collected from clinical trials, observational studies, case reports, and review literature. KEY FINDINGS COVID-19 can cause a spectrum of liver injuries, from mild enzyme elevations to severe hepatic dysfunction. Injury mechanisms include direct viral invasion, immune response alterations, drug toxicity, and hypoxia-reperfusion injury. Patients with chronic liver conditions (such as alcohol-related liver disease, nonalcoholic fatty liver disease, cirrhosis, and hepatocellular carcinoma) face increased risks of severe outcomes. The pandemic has worsened pre-existing liver conditions, disrupted cancer treatments, and complicated liver transplantation. Vaccination remains crucial for reducing severe disease, particularly in chronic liver patients and transplant recipients. Telemedicine has been beneficial in managing patients and reducing cross-infection risks. SIGNIFICANCE This review discusses the importance of improved diagnostic methods and management strategies for liver injury caused by COVID-19. It emphasizes the need for close monitoring and customized treatment for high-risk groups, advocating for future research to explore long-term effects, novel therapies, and evidence-based approaches to improve liver health during and after the pandemic.
Collapse
Affiliation(s)
- M Vinutha
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - Uday Raj Sharma
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India.
| | - Gurubasvaraja Swamy
- Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - S Rohini
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - Surendra Vada
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - Suresh Janandri
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - T Haribabu
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - Nageena Taj
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - S V Gayathri
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - S K Jyotsna
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - Manjunatha P Mudagal
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| |
Collapse
|
49
|
Tanoue K, Ohmura H, Uehara K, Ito M, Yamaguchi K, Tsuchihashi K, Shinohara Y, Lu P, Tamura S, Shimokawa H, Isobe T, Ariyama H, Shibata Y, Tanaka R, Kusaba H, Esaki T, Mitsugi K, Kiyozawa D, Iwasaki T, Yamamoto H, Oda Y, Akashi K, Baba E. Spatial dynamics of CD39 +CD8 + exhausted T cell reveal tertiary lymphoid structures-mediated response to PD-1 blockade in esophageal cancer. Nat Commun 2024; 15:9033. [PMID: 39426955 PMCID: PMC11490492 DOI: 10.1038/s41467-024-53262-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: 12/13/2023] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
Despite the success of immune checkpoint blockade (ICB) therapy for esophageal squamous cell cancer, the key immune cell populations that affect ICB efficacy remain unclear. Here, imaging mass cytometry of tumor tissues from ICB-treated patients identifies a distinct cell population of CD39+PD-1+CD8+ T cells, specifically the TCF1+ subset, precursor exhausted T (CD39+ Tpex) cells, which positively correlate with ICB benefit. CD39+ Tpex cells are predominantly in the stroma, while differentiated CD39+ exhausted T cells are abundantly and proximally within the parenchyma. Notably, CD39+ Tpex cells are concentrated within and around tertiary lymphoid structure (TLS). Accordingly, tumors harboring TLSs have more of these cells in tumor areas than tumors lacking TLSs, suggesting Tpex cell recruitment from TLSs to tumors. In addition, circulating CD39+ Tpex cells are also increased in responders following ICB therapy. Our findings show that this unique subpopulation of CD39+PD-1+CD8+ T cells is crucial for ICB benefit, and suggest a key role in TLS-mediated immune responses against tumors.
Collapse
Affiliation(s)
- Kenro Tanoue
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirofumi Ohmura
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koki Uehara
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mamoru Ito
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoko Yamaguchi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenji Tsuchihashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yudai Shinohara
- Department of Hematology/Oncology, Japan Community Healthcare Organization Kyushu Hospital, Fukuoka, Japan
| | - Peng Lu
- Department of Imaging Science Program, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Shingo Tamura
- Department of Medical Oncology, NHO National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Hozumi Shimokawa
- Department of Medical Oncology, Hamanomachi Hospital, Fukuoka, Japan
| | - Taichi Isobe
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroshi Ariyama
- Department of Medical Oncology, Kitakyushu Municipal Medical Center, Fukuoka, Japan
| | - Yoshihiro Shibata
- Department of Medical Oncology, Fukuoka Wajiro Hospital, Fukuoka, Japan
| | - Risa Tanaka
- Department of Medical Oncology, St Mary's Hospital, Kurume, Japan
| | - Hitoshi Kusaba
- Department of Medical Oncology, Hamanomachi Hospital, Fukuoka, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, National Kyushu Cancer Center, Fukuoka, Japan
| | - Kenji Mitsugi
- Department of Medical Oncology, Sasebo Kyosai Hospital, Nagasaki, Japan
| | - Daisuke Kiyozawa
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Iwasaki
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Pathology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eishi Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
50
|
Gottlieb RL, Clement M, Cook P, Deveikis A, Foong KS, Robinson P, Slim J, Spak CW, Buelens A, Callewaert K, De Meyer S, Mo WL, Verbrugge I, Van Wesenbeeck L, Zhuang Y, Chien JW, Opsomer M, Van Landuyt E. The IL-6 hypothesis in COVID-19: A phase 2, randomised, double-blind, placebo-controlled study to evaluate the efficacy and safety of free IL-6 sequestration by the monoclonal antibody sirukumab in severe and critical COVID-19. J Infect 2024; 89:106241. [PMID: 39182655 DOI: 10.1016/j.jinf.2024.106241] [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: 02/20/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Upregulation of IL-6 has been associated with worse prognosis in COVID-19 patients. Impact on IL-6 signalling has mostly been limited to clinical outcomes in IL-6 receptor antagonist trials. METHODS We performed a phase 2, randomised, double-blind, placebo-controlled trial (NCT04380961) of US-based hospitalised adults (<85 years) with laboratory-confirmed SARS-CoV-2 infection and severe (low levels of supplemental oxygen) or critical disease (high levels of oxygen supplementation). Patients received sirukumab 5 mg/kg or placebo single dose IV on Day 1 plus standard of care. The primary endpoint was time to sustained clinical improvement up to Day 28 based on an ordinal scale. Secondary endpoints included clinical improvement, all-cause mortality, and safety. Following an interim analysis, the protocol was amended to only recruit patients with critical COVID-19. FINDINGS From May 2020 to March 2021, 209 patients were randomised; 112 had critical disease (72 sirukumab, 40 placebo) at baseline. Median time to sustained clinical improvement in critical patients was 17 and 23 days in the sirukumab and placebo groups (HR, 1∙1; 95% CI, 0∙66-1∙88; p > 0∙05). At Day 28, 59∙4% versus 55∙0% of patients achieved clinical improvement with sirukumab versus placebo and rates of all-cause mortality were 24∙6% versus 30∙0%, respectively. Rates of grade ≥3 adverse events were comparable between the sirukumab and placebo groups (25∙9% vs 32∙9%; all patients). INTERPRETATION In critical COVID-19 patients who received sirukumab, there was no statistically significant difference in time to sustained clinical improvement versus placebo despite objective sequestration of circulating IL-6, questioning IL-6 as a key therapeutic target in COVID-19.
Collapse
Affiliation(s)
- Robert L Gottlieb
- Baylor University Medical Center, Dallas, TX, USA; Baylor Scott & White Research Institute, Dallas, TX, USA; Department of Internal Medicine, Burnett School of Medicine at TCU, Fort Worth, TX, USA; Department of Internal Medicine, Texas A&M Health Science Center, Dallas, TX, USA
| | - Meredith Clement
- Division of Infectious Diseases, Louisiana State University Health Sciences Center, New Orleans, LA, USA; University Medical Center, New Orleans, LA, USA
| | - Paul Cook
- Division of Infectious Diseases, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Audra Deveikis
- Bickerstaff Family Center at Miller Children's Hospital and Long Beach Memorial Medical Center, Long Beach, CA, USA
| | - Kap Sum Foong
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, MA, USA
| | | | - Jihad Slim
- Department of Internal Medicine, New York Medical College, Valhalla, NY, USA
| | - Cedric W Spak
- Baylor University Medical Center, Dallas, TX, USA; Department of Internal Medicine, Burnett School of Medicine at TCU, Fort Worth, TX, USA; Department of Internal Medicine, Texas A&M Health Science Center, Dallas, TX, USA; Baylor Scott & White Medical Center - All Saints, Fort Worth, TX, USA
| | | | | | | | | | | | | | - Yanli Zhuang
- Janssen Research & Development, LLC, Horsham, PA, USA
| | | | | | | |
Collapse
|