951
|
Tang J, Ravichandran S, Lee Y, Grubbs G, Coyle EM, Klenow L, Genser H, Golding H, Khurana S. Antibody affinity maturation and plasma IgA associate with clinical outcome in hospitalized COVID-19 patients. Nat Commun 2021; 12:1221. [PMID: 33619281 PMCID: PMC7900119 DOI: 10.1038/s41467-021-21463-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/27/2021] [Indexed: 01/10/2023] Open
Abstract
Hospitalized COVID-19 patients often present with a large spectrum of clinical symptoms. There is a critical need to better understand the immune responses to SARS-CoV-2 that lead to either resolution or exacerbation of the clinical disease. Here, we examine longitudinal plasma samples from hospitalized COVID-19 patients with differential clinical outcome. We perform immune-repertoire analysis including cytokine, hACE2-receptor inhibition, neutralization titers, antibody epitope repertoire, antibody kinetics, antibody isotype and antibody affinity maturation against the SARS-CoV-2 prefusion spike protein. Fatal cases demonstrate high plasma levels of IL-6, IL-8, TNFα, and MCP-1, and sustained high percentage of IgA-binding antibodies to prefusion spike compared with non-ICU survivors. Disease resolution in non-ICU and ICU patients associates with antibody binding to the receptor binding motif and fusion peptide, and antibody affinity maturation to SARS-CoV-2 prefusion spike protein. Here, we provide insight into the immune parameters associated with clinical disease severity and disease-resolution outcome in hospitalized patients that could inform development of vaccine/therapeutics against COVID-19. SARS-CoV2 infection has been linked to a wide range of clinical severities and the immunopathology is still under intense scrutiny. Here, the authors uncover an association of antibody affinity maturation and plasma IgA levels with clinical outcome in patients with COVID-19 disease.
Collapse
Affiliation(s)
- Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Supriya Ravichandran
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Youri Lee
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Elizabeth M Coyle
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Laura Klenow
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Hollie Genser
- Quest Diagnostics at Adventist Healthcare, Rockville, MD, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA.
| |
Collapse
|
952
|
Tomić S, Đokić J, Stevanović D, Ilić N, Gruden-Movsesijan A, Dinić M, Radojević D, Bekić M, Mitrović N, Tomašević R, Mikić D, Stojanović D, Čolić M. Reduced Expression of Autophagy Markers and Expansion of Myeloid-Derived Suppressor Cells Correlate With Poor T Cell Response in Severe COVID-19 Patients. Front Immunol 2021; 12:614599. [PMID: 33692788 PMCID: PMC7937809 DOI: 10.3389/fimmu.2021.614599] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
Widespread coronavirus disease (COVID)-19 is causing pneumonia, respiratory and multiorgan failure in susceptible individuals. Dysregulated immune response marks severe COVID-19, but the immunological mechanisms driving COVID-19 pathogenesis are still largely unknown, which is hampering the development of efficient treatments. Here we analyzed ~140 parameters of cellular and humoral immune response in peripheral blood of 41 COVID-19 patients and 16 age/gender-matched healthy donors by flow-cytometry, quantitative PCR, western blot and ELISA, followed by integrated correlation analyses with ~30 common clinical and laboratory parameters. We found that lymphocytopenia in severe COVID-19 patients (n=20) strongly affects T, NK and NKT cells, but not B cells and antibody production. Unlike increased activation of ICOS-1+ CD4+ T cells in mild COVID-19 patients (n=21), T cells in severe patients showed impaired activation, low IFN-γ production and high functional exhaustion, which correlated with significantly down-regulated HLA-DR expression in monocytes, dendritic cells and B cells. The latter phenomenon was followed by lower interferon responsive factor (IRF)-8 and autophagy-related genes expressions, and the expansion of myeloid derived suppressor cells (MDSC). Intriguingly, PD-L1-, ILT-3-, and IDO-1-expressing monocytic MDSC were the dominant producers of IL-6 and IL-10, which correlated with the increased inflammation and accumulation of regulatory B and T cell subsets in severe COVID-19 patients. Overall, down-regulated IRF-8 and autophagy-related genes expression, and the expansion of MDSC subsets could play critical roles in dysregulating T cell response in COVID-19, which could have large implications in diagnostics and design of novel therapeutics for this disease.
Collapse
Affiliation(s)
- Sergej Tomić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Jelena Đokić
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Dejan Stevanović
- Clinical Hospital Center Zemun, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nataša Ilić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Alisa Gruden-Movsesijan
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Miroslav Dinić
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Dušan Radojević
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marina Bekić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Nebojša Mitrović
- Clinical Hospital Center Zemun, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ratko Tomašević
- Clinical Hospital Center Zemun, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragan Mikić
- Clinics for Infectious and Tropical Diseases, Military Medical Academy, Belgrade, Serbia
| | - Dragoš Stojanović
- Clinical Hospital Center Zemun, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Miodrag Čolić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
- Department for Medical Sciences, Serbian Academy of Sciences and Arts, Belgrade, Serbia
| |
Collapse
|
953
|
Ziegler CGK, Miao VN, Owings AH, Navia AW, Tang Y, Bromley JD, Lotfy P, Sloan M, Laird H, Williams HB, George M, Drake RS, Christian T, Parker A, Sindel CB, Burger MW, Pride Y, Hasan M, Abraham GE, Senitko M, Robinson TO, Shalek AK, Glover SC, Horwitz BH, Ordovas-Montanes J. Impaired local intrinsic immunity to SARS-CoV-2 infection in severe COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.02.20.431155. [PMID: 33619488 PMCID: PMC7899452 DOI: 10.1101/2021.02.20.431155] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Infection with SARS-CoV-2, the virus that causes COVID-19, can lead to severe lower respiratory illness including pneumonia and acute respiratory distress syndrome, which can result in profound morbidity and mortality. However, many infected individuals are either asymptomatic or have isolated upper respiratory symptoms, which suggests that the upper airways represent the initial site of viral infection, and that some individuals are able to largely constrain viral pathology to the nasal and oropharyngeal tissues. Which cell types in the human nasopharynx are the primary targets of SARS-CoV-2 infection, and how infection influences the cellular organization of the respiratory epithelium remains incompletely understood. Here, we present nasopharyngeal samples from a cohort of 35 individuals with COVID-19, representing a wide spectrum of disease states from ambulatory to critically ill, as well as 23 healthy and intubated patients without COVID-19. Using standard nasopharyngeal swabs, we collected viable cells and performed single-cell RNA-sequencing (scRNA-seq), simultaneously profiling both host and viral RNA. We find that following infection with SARS-CoV-2, the upper respiratory epithelium undergoes massive reorganization: secretory cells diversify and expand, and mature epithelial cells are preferentially lost. Further, we observe evidence for deuterosomal cell and immature ciliated cell expansion, potentially representing active repopulation of lost ciliated cells through coupled secretory cell differentiation. Epithelial cells from participants with mild/moderate COVID-19 show extensive induction of genes associated with anti-viral and type I interferon responses. In contrast, cells from participants with severe lower respiratory symptoms appear globally muted in their anti-viral capacity, despite substantially higher local inflammatory myeloid populations and equivalent nasal viral loads. This suggests an essential role for intrinsic, local epithelial immunity in curbing and constraining viral-induced pathology. Using a custom computational pipeline, we characterized cell-associated SARS-CoV-2 RNA and identified rare cells with RNA intermediates strongly suggestive of active replication. Both within and across individuals, we find remarkable diversity and heterogeneity among SARS-CoV-2 RNA+ host cells, including developing/immature and interferon-responsive ciliated cells, KRT13+ "hillock"-like cells, and unique subsets of secretory, goblet, and squamous cells. Finally, SARS-CoV-2 RNA+ cells, as compared to uninfected bystanders, are enriched for genes involved in susceptibility (e.g., CTSL, TMPRSS2) or response (e.g., MX1, IFITM3, EIF2AK2) to infection. Together, this work defines both protective and detrimental host responses to SARS-CoV-2, determines the direct viral targets of infection, and suggests that failed anti-viral epithelial immunity in the nasal mucosa may underlie the progression to severe COVID-19.
Collapse
Affiliation(s)
- Carly G. K. Ziegler
- Program in Health Sciences & Technology, Harvard Medical School & MIT, Boston, MA 02115, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Vincent N. Miao
- Program in Health Sciences & Technology, Harvard Medical School & MIT, Boston, MA 02115, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anna H. Owings
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Andrew W. Navia
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ying Tang
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Joshua D. Bromley
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Microbiology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter Lotfy
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Meredith Sloan
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Hannah Laird
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Haley B. Williams
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Micayla George
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Riley S. Drake
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Taylor Christian
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Adam Parker
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Campbell B. Sindel
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Molly W. Burger
- Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Yilianys Pride
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Mohammad Hasan
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - George E. Abraham
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Michal Senitko
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Tanya O. Robinson
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Alex K. Shalek
- Program in Health Sciences & Technology, Harvard Medical School & MIT, Boston, MA 02115, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Sarah C. Glover
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bruce H. Horwitz
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Emergency Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Jose Ordovas-Montanes
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
954
|
Immune transcriptomes of highly exposed SARS-CoV-2 asymptomatic seropositive versus seronegative individuals from the Ischgl community. Sci Rep 2021; 11:4243. [PMID: 33608566 PMCID: PMC7895922 DOI: 10.1038/s41598-021-83110-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/28/2021] [Indexed: 01/07/2023] Open
Abstract
SARS-CoV-2 infection ranges from asymptomatic to severe with lingering symptomatology in some. This prompted investigation of whether or not asymptomatic disease results in measurable immune activation post-infection. Immune activation following asymptomatic SARS-CoV-2 infection was characterized through a comparative investigation of the immune cell transcriptomes from 43 asymptomatic seropositive and 52 highly exposed seronegative individuals from the same community 4–6 weeks following a superspreading event. Few of the 95 individuals had underlying health issues. One seropositive individual reported Cystic Fibrosis and one individual reported Incontinentia pigmenti. No evidence of immune activation was found in asymptomatic seropositive individuals with the exception of the Cystic Fibrosis patient. There were no statistically significant differences in immune transcriptomes between asymptomatic seropositive and highly exposed seronegative individuals. Four positive controls, mildly symptomatic seropositive individuals whose blood was examined 3 weeks following infection, showed immune activation. Negative controls were four seronegative individuals from neighboring communities without COVID-19. All individuals remained in their usual state of health through a five-month follow-up after sample collection. In summary, whole blood transcriptomes identified individual immune profiles within a community population and showed that asymptomatic infection within a super-spreading event was not associated with enduring immunological activation.
Collapse
|
955
|
Sette A, Crotty S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 2021; 184:861-880. [PMID: 33497610 PMCID: PMC7803150 DOI: 10.1016/j.cell.2021.01.007] [Citation(s) in RCA: 1276] [Impact Index Per Article: 319.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/28/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022]
Abstract
The adaptive immune system is important for control of most viral infections. The three fundamental components of the adaptive immune system are B cells (the source of antibodies), CD4+ T cells, and CD8+ T cells. The armamentarium of B cells, CD4+ T cells, and CD8+ T cells has differing roles in different viral infections and in vaccines, and thus it is critical to directly study adaptive immunity to SARS-CoV-2 to understand COVID-19. Knowledge is now available on relationships between antigen-specific immune responses and SARS-CoV-2 infection. Although more studies are needed, a picture has begun to emerge that reveals that CD4+ T cells, CD8+ T cells, and neutralizing antibodies all contribute to control of SARS-CoV-2 in both non-hospitalized and hospitalized cases of COVID-19. The specific functions and kinetics of these adaptive immune responses are discussed, as well as their interplay with innate immunity and implications for COVID-19 vaccines and immune memory against re-infection.
Collapse
Affiliation(s)
- Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| |
Collapse
|
956
|
Gonçalves de Andrade E, Šimončičová E, Carrier M, Vecchiarelli HA, Robert MÈ, Tremblay MÈ. Microglia Fighting for Neurological and Mental Health: On the Central Nervous System Frontline of COVID-19 Pandemic. Front Cell Neurosci 2021; 15:647378. [PMID: 33737867 PMCID: PMC7961561 DOI: 10.3389/fncel.2021.647378] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is marked by cardio-respiratory alterations, with increasing reports also indicating neurological and psychiatric symptoms in infected individuals. During COVID-19 pathology, the central nervous system (CNS) is possibly affected by direct severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) invasion, exaggerated systemic inflammatory responses, or hypoxia. Psychosocial stress imposed by the pandemic further affects the CNS of COVID-19 patients, but also the non-infected population, potentially contributing to the emergence or exacerbation of various neurological or mental health disorders. Microglia are central players of the CNS homeostasis maintenance and inflammatory response that exert their crucial functions in coordination with other CNS cells. During homeostatic challenges to the brain parenchyma, microglia modify their density, morphology, and molecular signature, resulting in the adjustment of their functions. In this review, we discuss how microglia may be involved in the neuroprotective and neurotoxic responses against CNS insults deriving from COVID-19. We examine how these responses may explain, at least partially, the neurological and psychiatric manifestations reported in COVID-19 patients and the general population. Furthermore, we consider how microglia might contribute to increased CNS vulnerability in certain groups, such as aged individuals and people with pre-existing conditions.
Collapse
Affiliation(s)
| | - Eva Šimončičová
- Division of Medical Science, University of Victoria, Victoria, BC, Canada
| | - Micaël Carrier
- Division of Medical Science, University of Victoria, Victoria, BC, Canada.,Axe Neurosciences, Centre de Recherche du CHU de Québec, Université de Laval, Québec City, QC, Canada
| | | | - Marie-Ève Robert
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université de Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Science, University of Victoria, Victoria, BC, Canada.,Axe Neurosciences, Centre de Recherche du CHU de Québec, Université de Laval, Québec City, QC, Canada.,Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada.,Department of Molecular Medicine, Université de Laval, Québec City, QC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
957
|
Wilson JK, Shankar-Hari M. Immunological Subpopulations Within Critically Ill COVID-19 Patients. Chest 2021; 159:1706-1708. [PMID: 33609517 PMCID: PMC7889441 DOI: 10.1016/j.chest.2021.01.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 01/08/2023] Open
Affiliation(s)
- Julie Kay Wilson
- School of Immunology & Microbial Sciences (J. Wilson and M. Shankar-Hari), Kings College London, London, England
| | - Manu Shankar-Hari
- School of Immunology & Microbial Sciences (J. Wilson and M. Shankar-Hari), Kings College London, London, England; Guy's and St Thomas' NHS Foundation Trust (M. Shankar-Hari), ICU Support Offices, St Thomas' Hospital, London, England.
| |
Collapse
|
958
|
Bonifacius A, Tischer-Zimmermann S, Dragon AC, Gussarow D, Vogel A, Krettek U, Gödecke N, Yilmaz M, Kraft ARM, Hoeper MM, Pink I, Schmidt JJ, Li Y, Welte T, Maecker-Kolhoff B, Martens J, Berger MM, Lobenwein C, Stankov MV, Cornberg M, David S, Behrens GMN, Witzke O, Blasczyk R, Eiz-Vesper B. COVID-19 immune signatures reveal stable antiviral T cell function despite declining humoral responses. Immunity 2021; 54:340-354.e6. [PMID: 33567252 PMCID: PMC7871825 DOI: 10.1016/j.immuni.2021.01.008] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/12/2020] [Accepted: 01/13/2021] [Indexed: 12/15/2022]
Abstract
Cellular and humoral immunity to SARS-CoV-2 is critical to control primary infection and correlates with severity of disease. The role of SARS-CoV-2-specific T cell immunity, its relationship to antibodies, and pre-existing immunity against endemic coronaviruses (huCoV), which has been hypothesized to be protective, were investigated in 82 healthy donors (HDs), 204 recovered (RCs), and 92 active COVID-19 patients (ACs). ACs had high amounts of anti-SARS-CoV-2 nucleocapsid and spike IgG but lymphopenia and overall reduced antiviral T cell responses due to the inflammatory milieu, expression of inhibitory molecules (PD-1, Tim-3) as well as effector caspase-3, -7, and -8 activity in T cells. SARS-CoV-2-specific T cell immunity conferred by polyfunctional, mainly interferon-γ-secreting CD4+ T cells remained stable throughout convalescence, whereas humoral responses declined. Immune responses toward huCoV in RCs with mild disease and strong cellular SARS-CoV-2 T cell reactivity imply a protective role of pre-existing immunity against huCoV.
Collapse
Affiliation(s)
- Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sabine Tischer-Zimmermann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Anna C Dragon
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Daniel Gussarow
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Alexander Vogel
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Ulrike Krettek
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Nina Gödecke
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | | | - Anke R M Kraft
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany; Centre for Individualised Infection Medicine (CiiM), Hannover, Germany
| | - Marius M Hoeper
- Department of Pneumology, Hannover Medical School, member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Isabell Pink
- Department of Pneumology, Hannover Medical School, member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Julius J Schmidt
- Department of Kidney and Hypertension Diseases, Hannover Medical School, Hannover, Germany
| | - Yang Li
- Department Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), Helmholtz Centre for Infection Research, Hannover Medical School, Hannover, Germany
| | - Tobias Welte
- Department of Pneumology, Hannover Medical School, member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Jörg Martens
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Marc Moritz Berger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, Essen, Germany
| | - Corinna Lobenwein
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Germany
| | - Metodi V Stankov
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany; Department of Rheumatology and Clinical Immunology, Hannover Medical School, Hannover, Germany
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany; Centre for Individualised Infection Medicine (CiiM), Hannover, Germany
| | - Sascha David
- Department of Kidney and Hypertension Diseases, Hannover Medical School, Hannover, Germany; Institute of Intensive Care Medicine, University Hospital Zurich, Switzerland
| | - Georg M N Behrens
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany; Department of Rheumatology and Clinical Immunology, Hannover Medical School, Hannover, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
959
|
Rha MS, Kim AR, Shin EC. SARS-CoV-2-Specific T Cell Responses in Patients with COVID-19 and Unexposed Individuals. Immune Netw 2021; 21:e2. [PMID: 33728095 PMCID: PMC7937509 DOI: 10.4110/in.2021.21.e2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes coronavirus disease 2019 (COVID-19), an ongoing pandemic disease. In the current review, we describe SARS-CoV-2-specific CD4+ and CD8+ T-cell responses in acute and convalescent COVID-19 patients. We also discuss the relationships between COVID-19 severity and SARS-CoV-2-specific T-cell responses and summarize recent reports regarding SARS-CoV-2-reactive T cells in SARS-CoV-2-unexposed individuals. These T cells may be cross-reactive cells primed by previous infection with human common-cold coronaviruses. Finally, we outline SARS-CoV-2-specific T-cell responses in the context of vaccination. A better understanding of SARS-CoV-2-specific T-cell responses is needed to develop effective vaccines and therapeutics.
Collapse
Affiliation(s)
- Min-Seok Rha
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - A Reum Kim
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Eui-Cheol Shin
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea.,The Center for Epidemic Preparedness, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
960
|
Alrubayyi A, Gea-Mallorquí E, Touizer E, Hameiri-Bowen D, Kopycinski J, Charlton B, Fisher-Pearson N, Muir L, Rosa A, Roustan C, Earl C, Cherepanov P, Pellegrino P, Waters L, Burns F, Kinloch S, Dong T, Dorrell L, Rowland-Jones S, McCoy LE, Peppa D. Characterization of humoral and SARS-CoV-2 specific T cell responses in people living with HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.02.15.431215. [PMID: 33619489 PMCID: PMC7899453 DOI: 10.1101/2021.02.15.431215] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
There is an urgent need to understand the nature of immune responses generated against SARS-CoV-2, to better inform risk-mitigation strategies for people living with HIV (PLWH). Although not all PLWH are considered immunosuppressed, residual cellular immune deficiency and ongoing inflammation could influence COVID-19 disease severity, the evolution and durability of protective memory responses. Here, we performed an integrated analysis, characterizing the nature, breadth and magnitude of SARS-CoV-2-specific immune responses in PLWH, controlled on ART, and HIV negative subjects. Both groups were in the convalescent phase of predominately mild COVID-19 disease. The majority of PLWH mounted SARS-CoV-2 Spike- and Nucleoprotein-specific antibodies with neutralizing activity and SARS-CoV-2-specific T cell responses, as measured by ELISpot, at levels comparable to HIV negative subjects. T cell responses against Spike, Membrane and Nucleocapsid were the most prominent, with SARS-CoV-2-specific CD4 T cells outnumbering CD8 T cells. Notably, the overall magnitude of SARS-CoV-2-specific T cell responses related to the size of the naive CD4 T cell pool and the CD4:CD8 ratio in PLWH, in whom disparate antibody and T cell responses were observed. Both humoral and cellular responses to SARS-CoV-2 were detected at 5-7 months post-infection, providing evidence of medium-term durability of responses irrespective of HIV serostatus. Incomplete immune reconstitution on ART and a low CD4:CD8 ratio could, however, hamper the development of immunity to SARS-CoV-2 and serve as a useful tool for risk stratification of PLWH. These findings have implications for the individual management and potential effectiveness of vaccination against SARS-CoV-2 in PLWH.
Collapse
Affiliation(s)
| | | | - Emma Touizer
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Dan Hameiri-Bowen
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
| | - Jakub Kopycinski
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
| | - Bethany Charlton
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
| | | | - Luke Muir
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Annachiara Rosa
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Chloe Roustan
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Christopher Earl
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Peter Cherepanov
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Pierre Pellegrino
- Mortimer Market Centre, Department of HIV, CNWL NHS Trust, London, United Kingdom
| | - Laura Waters
- Mortimer Market Centre, Department of HIV, CNWL NHS Trust, London, United Kingdom
| | - Fiona Burns
- Institute for Global Health UCL, London, United Kingdom
- Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Sabine Kinloch
- Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Tao Dong
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
| | - Lucy Dorrell
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
| | | | - Laura E. McCoy
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Dimitra Peppa
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
- Mortimer Market Centre, Department of HIV, CNWL NHS Trust, London, United Kingdom
| |
Collapse
|
961
|
Turnbull IR, Fuchs A, Remy KE, Kelly MP, Frazier EP, Ghosh S, Chang SW, Mazer M, Hess A, Leonard J, Hoofnagle M, Colonna M, Hotchkiss RS. Dysregulation of the Leukocyte Signaling Landscape during Acute COVID-19. RESEARCH SQUARE 2021:rs.3.rs-244150. [PMID: 33619472 PMCID: PMC7899466 DOI: 10.21203/rs.3.rs-244150/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The global COVID-19 pandemic has claimed the lives of more than 450,000 US citizens. Dysregulation of the immune system underlies the pathogenesis of COVID-19, with inflammation mediated local tissue injury to the lung in the setting of suppressed systemic immune function. To define the molecular mechanisms of immune dysfunction in COVID-19 we utilized a systems immunology approach centered on the circulating leukocyte phosphoproteome measured by mass cytometry. COVID-19 is associated with wholesale activation of a broad set of signaling pathways across myeloid and lymphoid cell populations. STAT3 phosphorylation predominated in both monocytes and T cells and was tightly correlated with circulating IL-6 levels. High levels of STAT3 phosphorylation was associated with decreased markers of myeloid cell maturation/activation and decreased ex-vivo T cell IFN-gamma production, demonstrating that during COVID-19 dysregulated cellular activation is associated with suppression of immune effector cell function. Collectively, these data reconcile the systemic inflammatory response and functional immunosuppression induced by COVID-19 and suggest STAT3 signaling may be the central pathophysiologic mechanism driving immune dysfunction in COVID-19.
Collapse
Affiliation(s)
- Isaiah R. Turnbull
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Anja Fuchs
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Kenneth E. Remy
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Michael P. Kelly
- Department of Orthopedic Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Elfaridah P. Frazier
- Department of Orthopedic Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Sarbani Ghosh
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Shin-Wen Chang
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Monty Mazer
- Department of Anesthesia, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Annie Hess
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Jennifer Leonard
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Mark Hoofnagle
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Richard S. Hotchkiss
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
962
|
Xie M, Yunis J, Yao Y, Shi J, Yang Y, Zhou P, Liang K, Wan Y, Mehdi A, Chen Z, Wang N, Xu S, Zhou M, Yu M, Wang K, Tao Y, Zhou Y, Li X, Liu X, Yu X, Wei Y, Liu Z, Sprent J, Yu D. High levels of soluble CD25 in COVID-19 severity suggest a divergence between anti-viral and pro-inflammatory T-cell responses. Clin Transl Immunology 2021; 10:e1251. [PMID: 33614032 PMCID: PMC7883478 DOI: 10.1002/cti2.1251] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Objectives We aimed to gain an understanding of the paradox of the immunity in COVID-19 patients with T cells showing both functional defects and hyperactivation and enhanced proliferation. Methods A total of 280 hospitalised patients with COVID-19 were evaluated for cytokine profiles and clinical features including viral shedding. A mouse model of acute infection by lymphocytic choriomeningitis virus (LCMV) was applied to dissect the relationship between immunological, virological and pathological features. The results from the mouse model were validated by published data set of single-cell RNA sequencing (scRNA-seq) of immune cells in bronchoalveolar lavage fluid (BALF) of COVID-19 patients. Results The levels of soluble CD25 (sCD25), IL-6, IL-8, IL-10 and TNF-α were higher in severe COVID-19 patients than non-severe cases, but only sCD25 was identified as an independent risk factor for disease severity by multivariable binary logistic regression analysis and showed a positive association with the duration of viral shedding. In agreement with the clinical observation, LCMV-infected mice with high levels of sCD25 demonstrated insufficient anti-viral response and delayed viral clearance. The elevation of sCD25 in mice was mainly contributed by the expansion of CD25+CD8+ T cells that also expressed the highest level of PD-1 with pro-inflammatory potential. The counterpart human CD25+PD-1+ T cells were expanded in BALF of COVID-19 patients with severe disease compared to those with modest disease. Conclusion These results suggest that high levels of sCD25 in COVID-19 patients probably result from insufficient anti-viral immunity and indicate an expansion of pro-inflammatory T cells that contribute to disease severity.
Collapse
Affiliation(s)
- Min Xie
- Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Respiratory Diseases National Ministry of Health of the People's Republic of China National Clinical Research Center for Respiratory Disease Wuhan China
| | - Joseph Yunis
- The University of Queensland Diamantina Institute Faculty of Medicine The University of Queensland Brisbane QLD Australia
| | - Yin Yao
- The University of Queensland Diamantina Institute Faculty of Medicine The University of Queensland Brisbane QLD Australia.,Department of Otolaryngology-Head and Neck Surgery Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jing Shi
- Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Respiratory Diseases National Ministry of Health of the People's Republic of China National Clinical Research Center for Respiratory Disease Wuhan China
| | - Yang Yang
- The University of Queensland Diamantina Institute Faculty of Medicine The University of Queensland Brisbane QLD Australia.,Shandong Artificial Intelligence Institute Qilu University of Technology (Shandong Academy of Sciences) Jinan China
| | - Pengcheng Zhou
- The University of Queensland Diamantina Institute Faculty of Medicine The University of Queensland Brisbane QLD Australia.,Department of Immunology and Infectious Disease John Curtin School of Medical Research Australian National University Canberra ACT Australia
| | - Kaili Liang
- China-Australia Centre for Personalised Immunology Shanghai Renji Hospital Shanghai Jiaotong University School of Medicine Shanghai
| | - Yanmin Wan
- Department of Infectious Diseases Huashan Hospital Fudan University Shanghai China.,Department of Radiology Shanghai Public Health Clinical Center Fudan University Shanghai China
| | - Ahmed Mehdi
- The University of Queensland Diamantina Institute Faculty of Medicine The University of Queensland Brisbane QLD Australia
| | - Zhian Chen
- The University of Queensland Diamantina Institute Faculty of Medicine The University of Queensland Brisbane QLD Australia
| | - Naiqi Wang
- The University of Queensland Diamantina Institute Faculty of Medicine The University of Queensland Brisbane QLD Australia
| | - Shuyun Xu
- Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Respiratory Diseases National Ministry of Health of the People's Republic of China National Clinical Research Center for Respiratory Disease Wuhan China
| | - Min Zhou
- Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Respiratory Diseases National Ministry of Health of the People's Republic of China National Clinical Research Center for Respiratory Disease Wuhan China
| | - Muqing Yu
- Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Respiratory Diseases National Ministry of Health of the People's Republic of China National Clinical Research Center for Respiratory Disease Wuhan China
| | - Ke Wang
- Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Respiratory Diseases National Ministry of Health of the People's Republic of China National Clinical Research Center for Respiratory Disease Wuhan China
| | - Yu Tao
- Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Respiratory Diseases National Ministry of Health of the People's Republic of China National Clinical Research Center for Respiratory Disease Wuhan China
| | - Ying Zhou
- Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Respiratory Diseases National Ministry of Health of the People's Republic of China National Clinical Research Center for Respiratory Disease Wuhan China
| | - Xiaochen Li
- Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Respiratory Diseases National Ministry of Health of the People's Republic of China National Clinical Research Center for Respiratory Disease Wuhan China
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Respiratory Diseases National Ministry of Health of the People's Republic of China National Clinical Research Center for Respiratory Disease Wuhan China
| | - Xiao Yu
- Department of Urology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yunbo Wei
- Shandong Artificial Intelligence Institute Qilu University of Technology (Shandong Academy of Sciences) Jinan China
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jonathan Sprent
- Immunology Division Garvan Institute of Medical Research Darlinghurst NSW Australia.,St Vincent's Clinical School University of New South Wales Sydney NSW Australia
| | - Di Yu
- The University of Queensland Diamantina Institute Faculty of Medicine The University of Queensland Brisbane QLD Australia.,Shandong Artificial Intelligence Institute Qilu University of Technology (Shandong Academy of Sciences) Jinan China.,Department of Immunology and Infectious Disease John Curtin School of Medical Research Australian National University Canberra ACT Australia.,China-Australia Centre for Personalised Immunology Shanghai Renji Hospital Shanghai Jiaotong University School of Medicine Shanghai
| |
Collapse
|
963
|
Bolouri H, Speake C, Skibinski D, Long SA, Hocking AM, Campbell DJ, Hamerman JA, Malhotra U, Buckner JH. The COVID-19 immune landscape is dynamically and reversibly correlated with disease severity. J Clin Invest 2021; 131:143648. [PMID: 33529167 DOI: 10.1172/jci143648] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/25/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUNDDespite a rapidly growing body of literature on coronavirus disease 2019 (COVID-19), our understanding of the immune correlates of disease severity, course, and outcome remains poor.METHODSUsing mass cytometry, we assessed the immune landscape in longitudinal whole-blood specimens from 59 patients presenting with acute COVID-19 and classified based on maximal disease severity. Hospitalized patients negative for SARS-CoV-2 were used as controls.RESULTSWe found that the immune landscape in COVID-19 formed 3 dominant clusters, which correlated with disease severity. Longitudinal analysis identified a pattern of productive innate and adaptive immune responses in individuals who had a moderate disease course, whereas those with severe disease had features suggestive of a protracted and dysregulated immune response. Further, we identified coordinate immune alterations accompanying clinical improvement and decline that were also seen in patients who received IL-6 pathway blockade.CONCLUSIONThe hospitalized COVID-19 negative cohort allowed us to identify immune alterations that were shared between severe COVID-19 and other critically ill patients. Collectively, our findings indicate that selection of immune interventions should be based in part on disease presentation and early disease trajectory due to the profound differences in the immune response in those with mild to moderate disease and those with the most severe disease.FUNDINGBenaroya Family Foundation, the Leonard and Norma Klorfine Foundation, Glenn and Mary Lynn Mounger, and the National Institutes of Health.
Collapse
Affiliation(s)
- Hamid Bolouri
- Center for Systems Immunology, Benaroya Research Institute (BRI) at Virginia Mason, Seattle, Washington, USA.,Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | | | | | | | - Daniel J Campbell
- Center for Fundamental Immunology, Benaroya Research Institute (BRI) at Virginia Mason, Seattle, Washington, USA.,Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jessica A Hamerman
- Center for Fundamental Immunology, Benaroya Research Institute (BRI) at Virginia Mason, Seattle, Washington, USA.,Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Uma Malhotra
- Department of Infectious Disease, Virginia Mason Medical Center, Seattle, Washington, USA.,Department of Medicine, Section of Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Jane H Buckner
- Center for Translational Immunology, and.,Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | | |
Collapse
|
964
|
Multisystem inflammatory syndrome in children during the COVID-19 pandemic in Turkey: first report from the Eastern Mediterranean. Clin Rheumatol 2021; 40:3227-3237. [PMID: 33576926 PMCID: PMC7879406 DOI: 10.1007/s10067-021-05631-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 11/04/2022]
Abstract
Objective We aimed to describe the typical clinical and laboratory features and treatment of children diagnosed with multisystem inflammatory syndrome in children (MIS-C) and to understand the differences as compared to severe/critical pediatric cases with COVID-19 in an eastern Mediterranean country. Methods Children (aged <18 years) who diagnosed with MIS-C and severe/critical pediatric cases with COVID-19 and were admitted to hospital between March 26 and November 3, 2020 were enrolled in the study. Results A total of 52 patients, 22 patients diagnosed with COVID-19 with severe/critical disease course and 30 patients diagnosed with MIS-C, were included in the study. Although severe COVID-19 cases and cases with MIS-C share many clinical and laboratory features, MIS-C cases had longer fever duration and higher rate of the existence of rash, conjunctival injection, peripheral edema, abdominal pain, altered mental status, and myalgia than in severe cases (p<0.001 for each). Of all, 53.3% of MIS-C cases had the evidence of myocardial involvement as compared to severe cases (27.2%). Additionally, C-reactive protein (CRP) and white blood cell (WBC) are the independent predictors for the diagnosis of MIS-C, particularly in the existence of conjunctival injection and rash. Corticosteroids, intravenous immunoglobulin (IVIG), and biologic immunomodulatory treatments were mainly used in MIS-C cases rather than cases with severe disease course. There were only three deaths among 52 patients, one of whom had Burkitt lymphoma and the two cases with severe COVID-19 of late referral. Conclusion Differences between clinical presentations, acute phase responses, organ involvements, and management strategies indicate that MIS-C might be a distinct immunopathogenic disease as compared to pediatric COVID-19. Conjunctival injection and higher CRP and low WBC count are reliable diagnostic parameters for MIS-C cases.Key Points • MIS-C cases had longer fever duration and higher rate of the existence of rash, conjunctival injection, peripheral edema, abdominal pain, altered mental status, and myalgia than in severe/critical pediatric cases with COVID-19. • Higher CRP and low total WBC count are the independent predictors for the diagnosis of MIS-C. • MIS-C might be a distinct immunopathogenic disease as compared to pediatric COVID-19. |
Collapse
|
965
|
Behm VAY, Bush CL. Personalized Nutrition 2020: Proceedings from the American Nutrition Association's 61st Annual Summit. J Am Coll Nutr 2021; 40:397-399. [PMID: 33576721 DOI: 10.1080/07315724.2021.1875340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The American Nutrition Association's 61st annual summit, Personalized Nutrition 2020, convened thought leaders and stakeholders to discuss innovations in personalized nutrition (PN) technology, science, and practice. Evolutions in PN science and technology are enabling novel inroads and applications, leading the ANA to launch a new component of its annual gathering, the Personalized Nutrition Business Leaders Forum. In light of renewed global interest in immune health during the COVID-19 pandemic, the flagship Science and Practice Conference focused on the topic of immune resilience. Presentations highlighted emerging research suggesting that individuals may have unique immunological responses to exogenous insults and that immune system resilience can be optimized by the application of nutritional factors that regulate immune function. Thus, PN tools and services may uniquely enhance immune preparedness by optimizing immune system function and status. Furthermore, PN practitioners trained to utilize emerging techniques and services can help prepare society to meet our modern immune challenges.HighlightsIn order to be effectively implemented, personalized nutrition requires ongoing research, innovative tools and services, and a specialized health care workforcePersonalized nutrition will continue to grow as an economic driver as consumer and patient interest surgeThere has been increased interest in the role of nutrition in immune function in light of COVID-19 and its comorbidities.
Collapse
Affiliation(s)
- Victoria A Y Behm
- American Nutrition Association, Hinsdale, Illinois, USA.,Nutrition and Herbal Medicine, Maryland University of Integrative Health, Laurel, Maryland, USA
| | | |
Collapse
|
966
|
Haslbauer JD, Matter MS, Stalder AK, Tzankov A. [Histomorphological patterns of regional lymph nodes in COVID-19 lungs]. DER PATHOLOGE 2021; 42:188-196. [PMID: 33575887 PMCID: PMC7877533 DOI: 10.1007/s00292-021-00914-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 12/07/2020] [Indexed: 01/08/2023]
Abstract
Hintergrund Eine dysregulierte Immunantwort, z. B. in der Form eines Zytokinsturmes, einer Störung des Immunglobulinklassenwechsels, eines sog. antikörpervermitteltem Enhancements oder einer aberranten Antigenpräsentation wurde bereits in schweren Krankheitsverläufen von COVID-19 beschrieben. Ziel der Arbeit Zur Charakterisierung der COVID-19-Immunantwort wurde die Histomorphologie der Lymphknoten des pulmonalen Abflussgebietes untersucht. Material und Methoden Regionale Lymphknoten des pulmonalen Abflussgebiets wurden bei COVID-19-Autopsien asserviert (n = 20). Deren Histomorphologie, SARS-CoV-2-qRT-PCR sowie Genexpressionsanalysen von gängigen Genen der Immunantwort wurden berücksichtigt. Ergebnisse Histologisch zeigten sich ein mäßig- bis schwergradiges Ödem mit Kapillarostase, eine erhöhte Anzahl von extrafollikulären Plasmablasten, milde bis mäßige Plasmazytose, vermehrte CD8+-T-Zellen und CD11c/CD68+-Histiozyten mit Hämophagozytoseaktivität. Von 20 Fällen wiesen 18 hypoplastische oder fehlende Keimzentren sowie eine Verminderung der follikulären dendritischen Zellen und follikulären T‑Helferzellen auf. In 14 von 20 Fällen war der qRT-PCR-Nachweis von SARS-CoV‑2 positiv, jedoch zeigte sich nur bei einem einzigen Fall eine immunhistochemische Positivität für SARS-CoV-2-N-Antigene in Sinushistiozyten. In Genexpressionsanalysen war eine erhöhte Expression von STAT1, CD163, Granzym B, CD8A, MZB1 und PAK1, neben CXCL9 zu beobachten. Diskussion Die Befunde in den Lymphknoten deuten auf eine dysregulierte Immunantwort bei schweren COVID-19-Krankheitsverläufen hin. Insbesondere impliziert das Ausbleiben der Keimzentrumsreaktion und die vermehrte Präsenz von Plasmablasten eine nur transiente B‑Zellreaktion, welche die Entwicklung einer Langzeitimmunität infrage stellt.
Collapse
Affiliation(s)
- Jasmin D Haslbauer
- Pathologie, Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Universität Basel, Basel, Schweiz
| | - Matthias S Matter
- Pathologie, Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Universität Basel, Basel, Schweiz
| | - Anna K Stalder
- Pathologie, Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Universität Basel, Basel, Schweiz
| | - Alexandar Tzankov
- Pathologie, Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Universität Basel, Basel, Schweiz. .,Institut für Medizinische Genetik und Pathologie, Schönbeinstrasse 40, 4031, Basel, Schweiz.
| |
Collapse
|
967
|
ElTanbouly MA, Zhao Y, Schaafsma E, Burns CM, Mabaera R, Cheng C, Noelle RJ. VISTA: A Target to Manage the Innate Cytokine Storm. Front Immunol 2021; 11:595950. [PMID: 33643285 PMCID: PMC7905033 DOI: 10.3389/fimmu.2020.595950] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, the success of immunotherapy targeting immunoregulatory receptors (immune checkpoints) in cancer have generated enthusiastic support to target these receptors in a wide range of other immune related diseases. While the overwhelming focus has been on blockade of these inhibitory pathways to augment immunity, agonistic triggering via these receptors offers the promise of dampening pathogenic inflammatory responses. V-domain Ig suppressor of T cell activation (VISTA) has emerged as an immunoregulatory receptor with constitutive expression on both the T cell and myeloid compartments, and whose agonistic targeting has proven a unique avenue relative to other checkpoint pathways to suppress pathologies mediated by the innate arm of the immune system. VISTA agonistic targeting profoundly changes the phenotype of human monocytes towards an anti-inflammatory cell state, as highlighted by striking suppression of the canonical markers CD14 and Fcγr3a (CD16), and the almost complete suppression of both the interferon I (IFN-I) and antigen presentation pathways. The insights from these very recent studies highlight the impact of VISTA agonistic targeting of myeloid cells, and its potential therapeutic implications in the settings of hyperinflammatory responses such as cytokine storms, driven by dysregulated immune responses to viral infections (with a focus on COVID-19) and autoimmune diseases. Collectively, these findings suggest that the VISTA pathway plays a conserved, non-redundant role in myeloid cell function.
Collapse
Affiliation(s)
- Mohamed A. ElTanbouly
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Yanding Zhao
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Evelien Schaafsma
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | | | - Rodwell Mabaera
- Department of Medicine, Norris Cotton Cancer Center, Lebanon, NH, United States
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, United States
| | - Randolph J. Noelle
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
968
|
Breton G, Mendoza P, Hägglöf T, Oliveira TY, Schaefer-Babajew D, Gaebler C, Turroja M, Hurley A, Caskey M, Nussenzweig MC. Persistent cellular immunity to SARS-CoV-2 infection. J Exp Med 2021; 218:211727. [PMID: 33533915 PMCID: PMC7845919 DOI: 10.1084/jem.20202515] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/21/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
SARS-CoV-2 is responsible for an ongoing pandemic that has affected millions of individuals around the globe. To gain further understanding of the immune response in recovered individuals, we measured T cell responses in paired samples obtained an average of 1.3 and 6.1 mo after infection from 41 individuals. The data indicate that recovered individuals show persistent polyfunctional SARS-CoV-2 antigen–specific memory that could contribute to rapid recall responses. Recovered individuals also show enduring alterations in relative overall numbers of CD4+ and CD8+ memory T cells, including expression of activation/exhaustion markers, and cell division.
Collapse
Affiliation(s)
- Gaëlle Breton
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Pilar Mendoza
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Thomas Hägglöf
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | | | - Christian Gaebler
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Martina Turroja
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Arlene Hurley
- Hospital Program Direction, The Rockefeller University, New York, NY
| | - Marina Caskey
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY.,Howard Hughes Medical Institute, Baltimore, MD
| |
Collapse
|
969
|
Cillo AR, Somasundaram A, Shan F, Cardello C, Workman CJ, Kitsios GD, Ruffin A, Kunning S, Lampenfeld C, Onkar S, Grebinoski S, Deshmukh G, Methe B, Liu C, Nambulli S, Andrews L, Duprex WP, Joglekar AV, Benos PV, Ray P, Ray A, McVerry BJ, Zhang Y, Lee JS, Das J, Singh H, Morris A, Bruno TC, Vignali DAA. Bifurcated monocyte states are predictive of mortality in severe COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.02.10.430499. [PMID: 33594364 PMCID: PMC7885916 DOI: 10.1101/2021.02.10.430499] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 infection presents with varied clinical manifestations1, ranging from mild symptoms to acute respiratory distress syndrome (ARDS) with high mortality2,3. Despite extensive analyses, there remains an urgent need to delineate immune cell states that contribute to mortality in severe COVID-19. We performed high-dimensional cellular and molecular profiling of blood and respiratory samples from critically ill COVID-19 patients to define immune cell genomic states that are predictive of outcome in severe COVID-19 disease. Critically ill patients admitted to the intensive care unit (ICU) manifested increased frequencies of inflammatory monocytes and plasmablasts that were also associated with ARDS not due to COVID-19. Single-cell RNAseq (scRNAseq)-based deconvolution of genomic states of peripheral immune cells revealed distinct gene modules that were associated with COVID-19 outcome. Notably, monocytes exhibited bifurcated genomic states, with expression of a cytokine gene module exemplified by CCL4 (MIP-1β) associated with survival and an interferon signaling module associated with death. These gene modules were correlated with higher levels of MIP-1β and CXCL10 levels in plasma, respectively. Monocytes expressing genes reflective of these divergent modules were also detectable in endotracheal aspirates. Machine learning algorithms identified the distinctive monocyte modules as part of a multivariate peripheral immune system state that was predictive of COVID-19 mortality. Follow-up analysis of the monocyte modules on ICU day 5 was consistent with bifurcated states that correlated with distinct inflammatory cytokines. Our data suggests a pivotal role for monocytes and their specific inflammatory genomic states in contributing to mortality in life-threatening COVID-19 disease and may facilitate discovery of new diagnostics and therapeutics.
Collapse
Affiliation(s)
- Anthony R Cillo
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
| | - Ashwin Somasundaram
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
| | - Feng Shan
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
- Integrative Systems Biology (ISB) Graduate Program, University of Pittsburgh School of Medicine, 200 Lothrop St., Pittsburgh, PA 15213, USA
| | - Carly Cardello
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
| | - Creg J Workman
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
| | - Georgios D Kitsios
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15213, USA
| | - Ayana Ruffin
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
- Graduate Program of Microbiology and Immunology (PMI), University of Pittsburgh School of Medicine, 200 Lothrop St., Pittsburgh, PA 15213, USA
| | - Sheryl Kunning
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
| | - Caleb Lampenfeld
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
| | - Sayali Onkar
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
- Graduate Program of Microbiology and Immunology (PMI), University of Pittsburgh School of Medicine, 200 Lothrop St., Pittsburgh, PA 15213, USA
| | - Stephanie Grebinoski
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
- Graduate Program of Microbiology and Immunology (PMI), University of Pittsburgh School of Medicine, 200 Lothrop St., Pittsburgh, PA 15213, USA
| | - Gaurav Deshmukh
- Meso Scale Discovery, A division of Meso Scale Diagnostics, LLC, 1601 Research Boulevard, Rockville, MD 20850-3173, USA
| | - Barbara Methe
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15213, USA
| | - Chang Liu
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
| | - Sham Nambulli
- Center for Vaccine Research, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lawrence Andrews
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
| | - W Paul Duprex
- Center for Vaccine Research, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alok V Joglekar
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Center for Systems Immunology, Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15213, USA
| | - Panayiotis V Benos
- Department of Computer Science, University of Pittsburgh, 4200 Fifth Avenue, Pittsburgh, PA 15260, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Prabir Ray
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- University of Pittsburgh Asthma Institute at the University of Pittsburgh Medical Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Anuradha Ray
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- University of Pittsburgh Asthma Institute at the University of Pittsburgh Medical Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Bryan J McVerry
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15213, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15213, USA
| | - Janet S Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jishnu Das
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Center for Systems Immunology, Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15213, USA
| | - Harinder Singh
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Center for Systems Immunology, Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15213, USA
| | - Alison Morris
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15213, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Dario A A Vignali
- Department of Immunology, School of Medicine, University of Pittsburgh. Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center. Pittsburgh, PA 15232, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
970
|
Feyaerts D, Hédou J, Gillard J, Chen H, Tsai ES, Peterson LS, Ando K, Manohar M, Do E, Dhondalay GK, Fitzpatrick J, Artandi M, Chang I, Snow TT, Chinthrajah RS, Warren CM, Wittman R, Meyerowitz JG, Ganio EA, Stelzer IA, Han X, Verdonk F, Gaudillière DK, Mukherjee N, Tsai AS, Rumer KK, Jiang S, Valdés Ferrer SI, Kelly JD, Furman D, Aghaeepour N, Angst MS, Boyd SD, Pinsky BA, Nolan GP, Nadeau KC, Gaudillière B, McIlwain DR. Integrated plasma proteomic and single-cell immune signaling network signatures demarcate mild, moderate, and severe COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.02.09.430269. [PMID: 33594362 PMCID: PMC7885914 DOI: 10.1101/2021.02.09.430269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The biological determinants of the wide spectrum of COVID-19 clinical manifestations are not fully understood. Here, over 1400 plasma proteins and 2600 single-cell immune features comprising cell phenotype, basal signaling activity, and signaling responses to inflammatory ligands were assessed in peripheral blood from patients with mild, moderate, and severe COVID-19, at the time of diagnosis. Using an integrated computational approach to analyze the combined plasma and single-cell proteomic data, we identified and independently validated a multivariate model classifying COVID-19 severity (multi-class AUCtraining = 0.799, p-value = 4.2e-6; multi-class AUCvalidation = 0.773, p-value = 7.7e-6). Features of this high-dimensional model recapitulated recent COVID-19 related observations of immune perturbations, and revealed novel biological signatures of severity, including the mobilization of elements of the renin-angiotensin system and primary hemostasis, as well as dysregulation of JAK/STAT, MAPK/mTOR, and NF-κB immune signaling networks. These results provide a set of early determinants of COVID-19 severity that may point to therapeutic targets for the prevention of COVID-19 progression.
Collapse
Affiliation(s)
- Dorien Feyaerts
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Julien Hédou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua Gillard
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, the Netherlands
| | - Han Chen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Eileen S. Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura S. Peterson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kazuo Ando
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Monali Manohar
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
| | - Evan Do
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
| | - Gopal K.R. Dhondalay
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
| | - Jessica Fitzpatrick
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
| | - Maja Artandi
- Department of Primary Care and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Iris Chang
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
| | - Theo T. Snow
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
| | - R. Sharon Chinthrajah
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Christopher M. Warren
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
| | - Rich Wittman
- Department of Primary Care and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Justin G. Meyerowitz
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Edward A. Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ina A. Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoyuan Han
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Sciences, University of the Pacific, Arthur A. Dugoni School of Dentistry, San Francisco, CA, USA
| | - Franck Verdonk
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Dyani K. Gaudillière
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Nilanjan Mukherjee
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Amy S. Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristen K. Rumer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sizun Jiang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sergio Iván Valdés Ferrer
- Departamento de Neurología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - J. Daniel Kelly
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, USA
- Institute of Global Health Sciences, UCSF, San Francisco, CA, USA
- F.I. Proctor Foundation, UCSF, San Francisco, CA, USA
| | - David Furman
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA
- Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA, USA
- Austral Institute for Applied Artificial Intelligence, Institute for Research in Translational Medicine (IIMT), Universidad Austral, CONICET, Pilar, Buenos Aires, Argentina
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Informatics, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Scott D. Boyd
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
| | - Benjamin A. Pinsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Garry P. Nolan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kari C. Nadeau
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - David R. McIlwain
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
971
|
Shi L, Huang H, Lu X, Yan X, Jiang X, Xu R, Wang S, Zhang C, Yuan X, Xu Z, Huang L, Fu JL, Li Y, Zhang Y, Yao WQ, Liu T, Song J, Sun L, Yang F, Zhang X, Zhang B, Shi M, Meng F, Song Y, Yu Y, Wen J, Li Q, Mao Q, Maeurer M, Zumla A, Yao C, Xie WF, Wang FS. Effect of human umbilical cord-derived mesenchymal stem cells on lung damage in severe COVID-19 patients: a randomized, double-blind, placebo-controlled phase 2 trial. Signal Transduct Target Ther 2021; 6:58. [PMID: 33568628 PMCID: PMC7873662 DOI: 10.1038/s41392-021-00488-5] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Treatment of severe Coronavirus Disease 2019 (COVID-19) is challenging. We performed a phase 2 trial to assess the efficacy and safety of human umbilical cord-mesenchymal stem cells (UC-MSCs) to treat severe COVID-19 patients with lung damage, based on our phase 1 data. In this randomized, double-blind, and placebo-controlled trial, we recruited 101 severe COVID-19 patients with lung damage. They were randomly assigned at a 2:1 ratio to receive either UC-MSCs (4 × 107 cells per infusion) or placebo on day 0, 3, and 6. The primary endpoint was an altered proportion of whole lung lesion volumes from baseline to day 28. Other imaging outcomes, 6-minute walk test (6-MWT), maximum vital capacity, diffusing capacity, and adverse events were recorded and analyzed. In all, 100 COVID-19 patients were finally received either UC-MSCs (n = 65) or placebo (n = 35). UC-MSCs administration exerted numerical improvement in whole lung lesion volume from baseline to day 28 compared with the placebo (the median difference was -13.31%, 95% CI -29.14%, 2.13%, P = 0.080). UC-MSCs significantly reduced the proportions of solid component lesion volume compared with the placebo (median difference: -15.45%; 95% CI -30.82%, -0.39%; P = 0.043). The 6-MWT showed an increased distance in patients treated with UC-MSCs (difference: 27.00 m; 95% CI 0.00, 57.00; P = 0.057). The incidence of adverse events was similar in the two groups. These results suggest that UC-MSCs treatment is a safe and potentially effective therapeutic approach for COVID-19 patients with lung damage. A phase 3 trial is required to evaluate effects on reducing mortality and preventing long-term pulmonary disability. (Funded by The National Key R&D Program of China and others. ClinicalTrials.gov number, NCT04288102.
Collapse
Affiliation(s)
- Lei Shi
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- Wuhan Huoshenshan Hospital, Wuhan, China
| | - Hai Huang
- Department of Respiratory, Changzheng Hospital, Second Military Medical University, Shanghai, China
- Optical Valley Branch of Maternal and Child Hospital of Hubei Province, Wuhan, China
| | - Xuechun Lu
- Wuhan Huoshenshan Hospital, Wuhan, China
- Department of Hematology, Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaoyan Yan
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China
| | - Xiaojing Jiang
- Department of Infectious Disease, General Hospital of Central Theater Command, Wuhan, China
| | - Ruonan Xu
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Siyu Wang
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Chao Zhang
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- Wuhan Huoshenshan Hospital, Wuhan, China
| | - Xin Yuan
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- Wuhan Huoshenshan Hospital, Wuhan, China
| | - Zhe Xu
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- Wuhan Huoshenshan Hospital, Wuhan, China
| | - Lei Huang
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- Wuhan Huoshenshan Hospital, Wuhan, China
| | - Jun-Liang Fu
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- Wuhan Huoshenshan Hospital, Wuhan, China
| | - Yuanyuan Li
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yu Zhang
- VCANBIO Cell & Gene Engineering Corp., Ltd, Tianjin, China
- National Industrial Base for Stem Cell Engineering Products, Tianjin, China
| | - Wei-Qi Yao
- National Industrial Base for Stem Cell Engineering Products, Tianjin, China
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyi Liu
- Wuhan Huoshenshan Hospital, Wuhan, China
- Key Laboratory of Cancer Center, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jinwen Song
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- Wuhan Huoshenshan Hospital, Wuhan, China
| | - Liangliang Sun
- Optical Valley Branch of Maternal and Child Hospital of Hubei Province, Wuhan, China
- Department of Endocrinology and Metabolism, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Fan Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zhang
- Wuhan Huoshenshan Hospital, Wuhan, China
- Nursing Department, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Bo Zhang
- Department of Infectious Disease, General Hospital of Central Theater Command, Wuhan, China
| | - Ming Shi
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Fanping Meng
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yanning Song
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yongpei Yu
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China
| | - Jiqiu Wen
- Wuhan Huoshenshan Hospital, Wuhan, China
| | - Qi Li
- Wuhan Huoshenshan Hospital, Wuhan, China
| | - Qing Mao
- Wuhan Huoshenshan Hospital, Wuhan, China
| | - Markus Maeurer
- Immunotherapy Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
- I Med Clinic, University of Mainz, Mainz, Germany
| | - Alimuddin Zumla
- Center for Clinical Microbiology, Division of Infection and Immunity, University College London, and UCL Hospitals NIHR Biomedical Research Centre, London, UK
| | - Chen Yao
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China.
| | - Wei-Fen Xie
- Optical Valley Branch of Maternal and Child Hospital of Hubei Province, Wuhan, China.
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, China.
| | - Fu-Sheng Wang
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.
- Wuhan Huoshenshan Hospital, Wuhan, China.
| |
Collapse
|
972
|
Abdul-Jawad S, Baù L, Alaguthurai T, Del Molino Del Barrio I, Laing AG, Hayday TS, Monin L, Muñoz-Ruiz M, McDonald L, Francos Quijorna I, McKenzie D, Davis R, Lorenc A, Chan JNE, Ryan S, Bugallo-Blanco E, Yorke R, Kamdar S, Fish M, Zlatareva I, Vantourout P, Jennings A, Gee S, Doores K, Bailey K, Hazell S, De Naurois J, Moss C, Russell B, Khan AA, Rowley M, Benjamin R, Enting D, Alrifai D, Wu Y, Zhou Y, Barber P, Ng T, Spicer J, Van Hemelrijck M, Kumar M, Vidler J, Lwin Y, Fields P, Karagiannis SN, Coolen ACC, Rigg A, Papa S, Hayday AC, Patten PEM, Irshad S. Acute Immune Signatures and Their Legacies in Severe Acute Respiratory Syndrome Coronavirus-2 Infected Cancer Patients. Cancer Cell 2021; 39:257-275.e6. [PMID: 33476581 PMCID: PMC7833668 DOI: 10.1016/j.ccell.2021.01.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/02/2020] [Accepted: 12/30/2020] [Indexed: 01/09/2023]
Abstract
Given the immune system's importance for cancer surveillance and treatment, we have investigated how it may be affected by SARS-CoV-2 infection of cancer patients. Across some heterogeneity in tumor type, stage, and treatment, virus-exposed solid cancer patients display a dominant impact of SARS-CoV-2, apparent from the resemblance of their immune signatures to those for COVID-19+ non-cancer patients. This is not the case for hematological malignancies, with virus-exposed patients collectively displaying heterogeneous humoral responses, an exhausted T cell phenotype and a high prevalence of prolonged virus shedding. Furthermore, while recovered solid cancer patients' immunophenotypes resemble those of non-virus-exposed cancer patients, recovered hematological cancer patients display distinct, lingering immunological legacies. Thus, while solid cancer patients, including those with advanced disease, seem no more at risk of SARS-CoV-2-associated immune dysregulation than the general population, hematological cancer patients show complex immunological consequences of SARS-CoV-2 exposure that might usefully inform their care.
Collapse
Affiliation(s)
- Sultan Abdul-Jawad
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Luca Baù
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Thanussuyah Alaguthurai
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK; Breast Cancer Now Research Unit, King's College London, London, UK
| | - Irene Del Molino Del Barrio
- Cancer Immunotherapy Accelerator, UCL Cancer Institute, University College and King's College, London, UK; Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Adam G Laing
- Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Thomas S Hayday
- Peter Gorer Department of Immunobiology, King's College London, London, UK
| | | | | | - Louisa McDonald
- Oncology and Haematology Clinical Trials (OHCT), Guy's and St Thomas' NHS Foundation Trust, London UK
| | | | | | - Richard Davis
- Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Anna Lorenc
- Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Julie Nuo En Chan
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Sarah Ryan
- Department of Inflammation Biology, King's College London, London, UK
| | - Eva Bugallo-Blanco
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Rozalyn Yorke
- Department of Inflammation Biology, King's College London, London, UK
| | - Shraddha Kamdar
- Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Matthew Fish
- Peter Gorer Department of Immunobiology, King's College London, London, UK; Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Iva Zlatareva
- Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Pierre Vantourout
- Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Aislinn Jennings
- Peter Gorer Department of Immunobiology, King's College London, London, UK; Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Sarah Gee
- Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Katie Doores
- Department of Infectious Diseases, King's College London, London, UK
| | - Katharine Bailey
- Department of Haematology, Guy's and St Thomas' NHS Foundation trust, London, UK
| | - Sophie Hazell
- Department of Haematology, Guy's and St Thomas' NHS Foundation trust, London, UK
| | - Julien De Naurois
- Department of Medical Oncology Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Charlotte Moss
- Department of Translational Oncology & Urology Research (TOUR), King's College London, London, UK
| | - Beth Russell
- Department of Translational Oncology & Urology Research (TOUR), King's College London, London, UK
| | - Aadil A Khan
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Mark Rowley
- London Institute for Mathematical Sciences, Mayfair, London, UK; Saddle Point Science Ltd, London, UK
| | - Reuben Benjamin
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK; Department of Haematological Medicine, King's College Hospital, London, UK
| | - Deborah Enting
- Department of Translational Oncology & Urology Research (TOUR), King's College London, London, UK
| | - Doraid Alrifai
- Department of Medical Oncology Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Yin Wu
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK; Cancer Immunotherapy Accelerator, UCL Cancer Institute, University College and King's College, London, UK; Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK
| | - You Zhou
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Paul Barber
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Tony Ng
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - James Spicer
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Mieke Van Hemelrijck
- Department of Translational Oncology & Urology Research (TOUR), King's College London, London, UK
| | - Mayur Kumar
- Department of Gastroenterology, Princess Royal University Hospital, Kent, UK
| | - Jennifer Vidler
- Department of Haematological Medicine, King's College Hospital, London, UK
| | - Yadanar Lwin
- Department of Haematological Medicine, King's College Hospital, London, UK
| | - Paul Fields
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK; Department of Haematology, Guy's and St Thomas' NHS Foundation trust, London, UK
| | - Sophia N Karagiannis
- Breast Cancer Now Research Unit, King's College London, London, UK; St. John's Institute of Dermatology, King's College London, London, UK; NIHR Biomedical Research Centre, and King's College London, London, UK
| | - Anthony C C Coolen
- Targeted Therapy Team, The Institute of Cancer Research, London, UK; London Institute for Mathematical Sciences, Mayfair, London, UK; Saddle Point Science Ltd, London, UK; Department of Biophysics, Radboud University, Nijmegen, The Netherlands
| | - Anne Rigg
- Department of Medical Oncology Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Sophie Papa
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK; Department of Medical Oncology Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Adrian C Hayday
- Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK
| | - Piers E M Patten
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK; Department of Haematological Medicine, King's College Hospital, London, UK; Medical Research Council (MRC) Clinical Academic Research Partnership, London, UK
| | - Sheeba Irshad
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK; Breast Cancer Now Research Unit, King's College London, London, UK; Department of Medical Oncology Guy's and St Thomas' NHS Foundation Trust, London, UK; Cancer Research UK (CRUK) Clinician Scientist, London, UK.
| |
Collapse
|
973
|
Rupp J, Dreo B, Gütl K, Fessler J, Moser A, Haditsch B, Schilcher G, Matzkies LM, Steinmetz I, Greinix H, Stradner MH. T Cell Phenotyping in Individuals Hospitalized with COVID-19. THE JOURNAL OF IMMUNOLOGY 2021; 206:1478-1482. [PMID: 33558375 DOI: 10.4049/jimmunol.2001034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/12/2021] [Indexed: 01/03/2023]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has become pandemic. Cytokine release syndrome occurring in a minority of SARS-CoV-2 infections is associated with severe disease and high mortality. We profiled the composition, activation, and proliferation of T cells in 20 patients with severe or critical COVID-19 and 40 matched healthy controls by flow cytometry. Unsupervised hierarchical cluster analysis based on 18 T cell subsets resulted in separation of healthy controls and COVID-19 patients. Compared to healthy controls, patients suffering from severe and critical COVID-19 had increased frequencies of activated and proliferating CD38+Ki67+ CD4+ and CD8+ T cells, suggesting active antiviral T cell defense. Frequencies of CD38+Ki67+ Th1 and CD4+ cells correlated negatively with plasma IL-6. Thus, our data suggest that patients suffering from COVID-19 have a distinct T cell composition that is potentially modulated by IL-6.
Collapse
Affiliation(s)
- Janine Rupp
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Barbara Dreo
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Katharina Gütl
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Johannes Fessler
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Adrian Moser
- Österreichische Gesundheitskasse, Gesundheitszentrum Graz, 8010 Graz, Austria
| | - Bernd Haditsch
- Österreichische Gesundheitskasse, Gesundheitszentrum Graz, 8010 Graz, Austria
| | - Gernot Schilcher
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Lucie-Marie Matzkies
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, 8036 Graz, Austria; and
| | - Ivo Steinmetz
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, 8036 Graz, Austria; and
| | - Hildegard Greinix
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Martin H Stradner
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria;
| |
Collapse
|
974
|
Neidleman J, Luo X, George AF, McGregor M, Yang J, Yun C, Murray V, Gill G, Greene WC, Vasquez J, Lee S, Ghosn E, Lynch K, Roan NR. Distinctive features of SARS-CoV-2-specific T cells predict recovery from severe COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.01.22.21250054. [PMID: 33532792 PMCID: PMC7852243 DOI: 10.1101/2021.01.22.21250054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Although T cells are likely players in SARS-CoV-2 immunity, little is known about the phenotypic features of SARS-CoV-2-specific T cells associated with recovery from severe COVID-19. We analyzed T cells from longitudinal specimens of 34 COVID-19 patients with severities ranging from mild (outpatient) to critical culminating in death. Relative to patients that succumbed, individuals that recovered from severe COVID-19 harbored elevated and increasing numbers of SARS-CoV-2-specific T cells capable of homeostatic proliferation. In contrast, fatal COVID-19 displayed elevated numbers of SARS-CoV-2-specific regulatory T cells and a time-dependent escalation in activated bystander CXCR4+ T cells. Together with the demonstration of increased proportions of inflammatory CXCR4+ T cells in the lungs of severe COVID-19 patients, these results support a model whereby lung-homing T cells activated through bystander effects contribute to immunopathology, while a robust, non-suppressive SARS-CoV-2-specific T cell response limits pathogenesis and promotes recovery from severe COVID-19.
Collapse
Affiliation(s)
- Jason Neidleman
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Xiaoyu Luo
- Gladstone Institutes, San Francisco, CA, USA
| | - Ashley F. George
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Matthew McGregor
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Junkai Yang
- Department of Medicine, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Cassandra Yun
- Department of Laboratory Medicine, University of California, San Francisco, CA USA
| | - Victoria Murray
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA, USA
| | - Gurjot Gill
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA, USA
| | - Warner C. Greene
- Gladstone Institutes, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Joshua Vasquez
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Sulggi Lee
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA, USA
| | - Eliver Ghosn
- Department of Medicine, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Kara Lynch
- Department of Laboratory Medicine, University of California, San Francisco, CA USA
| | - Nadia R. Roan
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| |
Collapse
|
975
|
Koutsakos M, Rowntree LC, Hensen L, Chua BY, van de Sandt CE, Habel JR, Zhang W, Jia X, Kedzierski L, Ashhurst TM, Putri GH, Marsh-Wakefield F, Read MN, Edwards DN, Clemens EB, Wong CY, Mordant FL, Juno JA, Amanat F, Audsley J, Holmes NE, Gordon CL, Smibert OC, Trubiano JA, Hughes CM, Catton M, Denholm JT, Tong SY, Doolan DL, Kotsimbos TC, Jackson DC, Krammer F, Godfrey DI, Chung AW, King NJ, Lewin SR, Wheatley AK, Kent SJ, Subbarao K, McMahon J, Thevarajan I, Nguyen TH, Cheng AC, Kedzierska K. Integrated immune dynamics define correlates of COVID-19 severity and antibody responses. CELL REPORTS MEDICINE 2021; 2:100208. [PMID: 33564749 PMCID: PMC7862905 DOI: 10.1016/j.xcrm.2021.100208] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/05/2020] [Accepted: 02/01/2021] [Indexed: 12/15/2022]
Abstract
SARS-CoV-2 causes a spectrum of COVID-19 disease, the immunological basis of which remains ill defined. We analyzed 85 SARS-CoV-2-infected individuals at acute and/or convalescent time points, up to 102 days after symptom onset, quantifying 184 immunological parameters. Acute COVID-19 presented with high levels of IL-6, IL-18, and IL-10 and broad activation marked by the upregulation of CD38 on innate and adaptive lymphocytes and myeloid cells. Importantly, activated CXCR3+cTFH1 cells in acute COVID-19 significantly correlate with and predict antibody levels and their avidity at convalescence as well as acute neutralization activity. Strikingly, intensive care unit (ICU) patients with severe COVID-19 display higher levels of soluble IL-6, IL-6R, and IL-18, and hyperactivation of innate, adaptive, and myeloid compartments than patients with moderate disease. Our analyses provide a comprehensive map of longitudinal immunological responses in COVID-19 patients and integrate key cellular pathways of complex immune networks underpinning severe COVID-19, providing important insights into potential biomarkers and immunotherapies. Analyses of 184 immune features define kinetics of immune responses to SARS-CoV-2 Circulating TFH1 cells in acute COVID-19 correlate with antibodies sIL-6R levels are elevated in severe COVID-19 but do not correlate with IL-6 Elevated IL-6 and IL-18 correlate with immune cell hyperactivation
Collapse
Affiliation(s)
- Marios Koutsakos
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Louise C. Rowntree
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Luca Hensen
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Brendon Y. Chua
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Carolien E. van de Sandt
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jennifer R. Habel
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Wuji Zhang
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Thomas M. Ashhurst
- Sydney Cytometry Core Research Facility, Charles Perkins Centre, Centenary Institute and University of Sydney, Sydney, NSW, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia
| | - Givanna H. Putri
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Computer Science, University of Sydney, Sydney, NSW, Australia
| | - Felix Marsh-Wakefield
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Vascular Immunology Unit, Discipline of Pathology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Mark N. Read
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Computer Science, University of Sydney, Sydney, NSW, Australia
- The Westmead Initiative, University of Sydney, Sydney, NSW, Australia
| | - Davis N. Edwards
- School of Computer Science, University of Sydney, Sydney, NSW, Australia
- The Westmead Initiative, University of Sydney, Sydney, NSW, Australia
| | - E. Bridie Clemens
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Chinn Yi Wong
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Francesca L. Mordant
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jennifer A. Juno
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | - Jennifer Audsley
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Natasha E. Holmes
- Department of Infectious Diseases, Austin Hospital, Heidelberg, VIC, Australia
- Department of Medicine and Radiology, University of Melbourne, Parkville, VIC, Australia
- Data Analytics Research and Evaluation (DARE) Centre, Austin Health and University of Melbourne, Heidelberg, VIC, Australia
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC, Australia
| | - Claire L. Gordon
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Infectious Diseases, Austin Hospital, Heidelberg, VIC, Australia
| | - Olivia C. Smibert
- Department of Infectious Diseases, Austin Hospital, Heidelberg, VIC, Australia
- Department of Infectious Diseases, Peter McCallum Cancer Centre, Melbourne, VIC, Australia
- National Centre for Infections in Cancer, Peter McCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jason A. Trubiano
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC, Australia
- Department of Infectious Diseases, Peter McCallum Cancer Centre, Melbourne, VIC, Australia
- National Centre for Infections in Cancer, Peter McCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Medicine (Austin Health), University of Melbourne, Heidelberg, VIC, Australia
| | - Carly M. Hughes
- Monash Infectious Diseases, Monash Medical Centre, Monash Health, Melbourne, VIC, Australia
| | - Mike Catton
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Justin T. Denholm
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Services, Royal Melbourne Hospital and Doherty Department University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC, Australia
| | - Steven Y.C. Tong
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Services, Royal Melbourne Hospital and Doherty Department University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC, Australia
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Denise L. Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - Tom C. Kotsimbos
- Department of Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Allergy, Immunology, and Respiratory Medicine, Alfred Hospital, Melbourne, VIC, Australia
| | - David C. Jackson
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging at the University of Melbourne, Melbourne, VIC, Australia
| | - Amy W. Chung
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Nicholas J.C. King
- Sydney Cytometry Core Research Facility, Charles Perkins Centre, Centenary Institute and University of Sydney, Sydney, NSW, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Viral Immunopathology Laboratory, Discipline of Pathology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Sydney Nano, University of Sydney, Sydney, NSW 2006, Australia
| | - Sharon R. Lewin
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Services, Royal Melbourne Hospital and Doherty Department University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC, Australia
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, VIC, Australia
| | - Adam K. Wheatley
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC, Australia
- Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- World Health Organization Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - James McMahon
- Monash Infectious Diseases, Monash Medical Centre, Monash Health, Melbourne, VIC, Australia
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, VIC, Australia
| | - Irani Thevarajan
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Services, Royal Melbourne Hospital and Doherty Department University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC, Australia
| | - Thi H.O. Nguyen
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Allen C. Cheng
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Infection Prevention and Healthcare Epidemiology Unit, Alfred Health, Melbourne, VIC, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
- Corresponding author
| |
Collapse
|
976
|
Deschler S, Kager J, Erber J, Fricke L, Koyumdzhieva P, Georgieva A, Lahmer T, Wiessner JR, Voit F, Schneider J, Horstmann J, Iakoubov R, Treiber M, Winter C, Ruland J, Busch DH, Knolle PA, Protzer U, Spinner CD, Schmid RM, Quante M, Böttcher K. Mucosal-Associated Invariant T (MAIT) Cells Are Highly Activated and Functionally Impaired in COVID-19 Patients. Viruses 2021; 13:241. [PMID: 33546489 PMCID: PMC7913667 DOI: 10.3390/v13020241] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), comprises mild courses of disease as well as progression to severe disease, characterised by lung and other organ failure. The immune system is considered to play a crucial role for the pathogenesis of COVID-19, although especially the contribution of innate-like T cells remains poorly understood. Here, we analysed the phenotype and function of mucosal-associated invariant T (MAIT) cells, innate-like T cells with potent antimicrobial effector function, in patients with mild and severe COVID-19 by multicolour flow cytometry. Our data indicate that MAIT cells are highly activated in patients with COVID-19, irrespective of the course of disease, and express high levels of proinflammatory cytokines such as IL-17A and TNFα ex vivo. Of note, expression of the activation marker HLA-DR positively correlated with SAPS II score, a measure of disease severity. Upon MAIT cell-specific in vitro stimulation, MAIT cells however failed to upregulate expression of the cytokines IL-17A and TNFα, as well as cytolytic proteins, that is, granzyme B and perforin. Thus, our data point towards an altered cytokine expression profile alongside an impaired antibacterial and antiviral function of MAIT cells in COVID-19 and thereby contribute to the understanding of COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
- Sebastian Deschler
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Juliane Kager
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Johanna Erber
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Lisa Fricke
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Plamena Koyumdzhieva
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Alexandra Georgieva
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Tobias Lahmer
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Johannes R. Wiessner
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Florian Voit
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Jochen Schneider
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Julia Horstmann
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Roman Iakoubov
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Matthias Treiber
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Christof Winter
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, 81675 Munich, Germany; (C.W.); (J.R.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, 81675 Munich, Germany; (C.W.); (J.R.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, 81675 Munich, Germany;
| | - Percy A. Knolle
- Institute of Molecular Immunology and Experimental Oncology, University Hospital Rechts der Isar, Technical University of Munich, 81675 Munich, Germany;
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, 81675 Munich, Germany;
- German Center for Infection Research (DZIF), 38124 Braunschweig, Partner Site Munich, Germany
| | - Christoph D. Spinner
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
- German Center for Infection Research (DZIF), 38124 Braunschweig, Partner Site Munich, Germany
| | - Roland M. Schmid
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| | - Michael Quante
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
- Freiburg University Medical Center, Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Katrin Böttcher
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; (S.D.); (J.K.); (J.E.); (L.F.); (P.K.); (A.G.); (T.L.); (J.R.W.); (F.V.); (J.S.); (J.H.); (R.I.); (M.T.); (C.D.S.); (R.M.S.); (M.Q.)
| |
Collapse
|
977
|
Bange EM, Han NA, Wileyto P, Kim JY, Gouma S, Robinson J, Greenplate AR, Porterfield F, Owoyemi O, Naik K, Zheng C, Galantino M, Weisman AR, Ittner CA, Kugler EM, Baxter AE, Oniyide O, Agyekum RS, Dunn TG, Jones TK, Giannini HM, Weirick ME, McAllister CM, Babady NE, Kumar A, Widman AJ, DeWolf S, Boutemine SR, Roberts C, Budzik KR, Tollett S, Wright C, Perloff T, Sun L, Mathew D, Giles JR, Oldridge DA, Wu JE, Alanio C, Adamski S, Garfall AL, Vella L, Kerr SJ, Cohen JV, Oyer RA, Massa R, Maillard IP, The UPenn COVID Processing Unit, Maxwell KN, Reilly JP, Maslak PG, Vonderheide RH, Wolchok JD, Hensley SE, Wherry EJ, Meyer N, DeMichele AM, Vardhana SA, Mamtani R, Huang AC. CD8 T cells compensate for impaired humoral immunity in COVID-19 patients with hematologic cancer. RESEARCH SQUARE 2021:rs.3.rs-162289. [PMID: 33564756 PMCID: PMC7872363 DOI: 10.21203/rs.3.rs-162289/v1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer patients have increased morbidity and mortality from Coronavirus Disease 2019 (COVID-19), but the underlying immune mechanisms are unknown. In a cohort of 100 cancer patients hospitalized for COVID-19 at the University of Pennsylvania Health System, we found that patients with hematologic cancers had a significantly higher mortality relative to patients with solid cancers after accounting for confounders including ECOG performance status and active cancer status. We performed flow cytometric and serologic analyses of 106 cancer patients and 113 non-cancer controls from two additional cohorts at Penn and Memorial Sloan Kettering Cancer Center. Patients with solid cancers exhibited an immune phenotype similar to non-cancer patients during acute COVID-19 whereas patients with hematologic cancers had significant impairment of B cells and SARS-CoV-2-specific antibody responses. High dimensional analysis of flow cytometric data revealed 5 distinct immune phenotypes. An immune phenotype characterized by CD8 T cell depletion was associated with a high viral load and the highest mortality of 71%, among all cancer patients. In contrast, despite impaired B cell responses, patients with hematologic cancers and preserved CD8 T cells had a lower viral load and mortality. These data highlight the importance of CD8 T cells in acute COVID-19, particularly in the setting of impaired humoral immunity. Further, depletion of B cells with anti-CD20 therapy resulted in almost complete abrogation of SARS-CoV-2-specific IgG and IgM antibodies, but was not associated with increased mortality compared to other hematologic cancers, when adequate CD8 T cells were present. Finally, higher CD8 T cell counts were associated with improved overall survival in patients with hematologic cancers. Thus, CD8 T cells likely compensate for deficient humoral immunity and influence clinical recovery of COVID-19. These observations have important implications for cancer and COVID-19-directed treatments, immunosuppressive therapies, and for understanding the role of B and T cells in acute COVID-19.
Collapse
Affiliation(s)
- Erin M. Bange
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Nicholas A. Han
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
| | - Paul Wileyto
- Abramson Cancer Center, University of Pennsylvania
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania
| | - Justin Y. Kim
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | | | - Allison R. Greenplate
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Florence Porterfield
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Olutosin Owoyemi
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Karan Naik
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Cathy Zheng
- Abramson Cancer Center, University of Pennsylvania
| | | | - Ariel R. Weisman
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Caroline A.G. Ittner
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Emily M. Kugler
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Amy E. Baxter
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Olutwatosin Oniyide
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Roseline S. Agyekum
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Thomas G. Dunn
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Tiffanie K. Jones
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Heather M. Giannini
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Madison E. Weirick
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | | | - N. Esther Babady
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| | - Anita Kumar
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Adam J Widman
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Susan DeWolf
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | | | | | | | | | - Carla Wright
- Abramson Cancer Center, University of Pennsylvania
| | - Tara Perloff
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital
| | - Lova Sun
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Divij Mathew
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Josephine R. Giles
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Derek A. Oldridge
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Jennifer E. Wu
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Cécile Alanio
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Sharon Adamski
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Alfred L. Garfall
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Laura Vella
- Department of Pediatrics, Perelman School of Medicine, Children’s Hospital of Philadelphia
| | - Samuel J. Kerr
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital
| | - Justine V. Cohen
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital
| | - Randall A. Oyer
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital
| | - Ryan Massa
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital
| | - Ivan P. Maillard
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | | | - Kara N. Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - John P. Reilly
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Peter G. Maslak
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| | - Robert H. Vonderheide
- Abramson Cancer Center, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Jedd D. Wolchok
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Scott E. Hensley
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | - E. John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Nuala Meyer
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Angela M. DeMichele
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Santosha A. Vardhana
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Parker Institute for Cancer Immunotherapy
| | - Ronac Mamtani
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Alexander C. Huang
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| |
Collapse
|
978
|
Pereira NL, Ahmad F, Byku M, Cummins NW, Morris AA, Owens A, Tuteja S, Cresci S. COVID-19: Understanding Inter-Individual Variability and Implications for Precision Medicine. Mayo Clin Proc 2021; 96:446-463. [PMID: 33549263 PMCID: PMC7713605 DOI: 10.1016/j.mayocp.2020.11.024] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/09/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is characterized by heterogeneity in susceptibility to the disease and severity of illness. Understanding inter-individual variation has important implications for not only allocation of resources but also targeting patients for escalation of care, inclusion in clinical trials, and individualized medical therapy including vaccination. In addition to geographic location and social vulnerability, there are clear biological differences such as age, sex, race, presence of comorbidities, underlying genetic variation, and differential immune response that contribute to variability in disease manifestation. These differences may have implications for precision medicine. Specific examples include the observation that androgens regulate the expression of the enzyme transmembrane protease, serine 2 which facilitates severe acute respiratory syndrome coronavirus 2 viral entry into the cell; therefore, androgen deprivation therapy is being explored as a treatment option in males infected with COVID-19. An immunophenotyping study of COVID-19 patients has shown that a subset develop T cytopenia which has prompted a clinical trial that is testing the efficacy of interleukin-7 in these patients. Predicting which COVID-19 patients will develop progressive disease that will require hospitalization has important implications for clinical trials that target outpatients. Enrollment of patients at low risk for progression of disease and hospitalization would likely not result in such therapy demonstrating efficacy. There are efforts to use artificial intelligence to integrate digital data from smartwatch applications or digital monitoring systems and biological data to enable identification of the high risk COVID-19 patient. The ultimate goal of precision medicine using such modern technology is to recognize individual differences to improve health for all.
Collapse
Affiliation(s)
- Naveen L Pereira
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN.
| | - Ferhaan Ahmad
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine Iowa City, IA
| | - Mirnela Byku
- Department of Medicine, Division of Cardiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | | | - Anjali Owens
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sony Tuteja
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sharon Cresci
- Department of Medicine and Genetics, Washington University, St Louis, MO
| |
Collapse
|
979
|
Bland JS. COVID-19 Risk: Clinical Tools for Assessing and Personalizing Immunity. Integr Med (Encinitas) 2021; 20:18-23. [PMID: 34393672 PMCID: PMC8352418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Researchers and clinicians all over the world are struggling to bring the spread of SARS-CoV-2 under control. Whether we refer to COVID-19 as a pandemic (a widely used word) or a syndemic (a new emerging term), we now know that specific immunotypes have been linked to both risk to infection and presentation of this disease. Application of assessment tools that support a fuller understanding of individual immune system status is next-level care that providers must prepare for and deliver. Commonly used biometric devices already gather data that can be relevant and useful to a phenotypic evaluation of immune function. These variables include pulse rate, blood oxygenation, sleep cycles, respiration rate, heart rate variability, continuous blood glucose monitoring, and ambulatory blood pressure. When coupled with traditional blood analytes and measurements of nutrient status, a more complete picture of immunological function may be revealed. Innovative questionnaires and algorithms can also be helpful additions to a clinician's toolkit. In a therapeutic relationship between provider and patient, this approach may lead to options for personalized immune intervention using diet, medical nutrition, and lifestyle medicine.
Collapse
|
980
|
Abstract
Vaccines are urgently needed to control the coronavirus disease 2019 (COVID-19) pandemic and to help the return to pre-pandemic normalcy. A great many vaccine candidates are being developed, several of which have completed late-stage clinical trials and are reporting positive results. In this Progress article, we discuss which viral elements are used in COVID-19 vaccine candidates, why they might act as good targets for the immune system and the implications for protective immunity.
Collapse
Affiliation(s)
- Lianpan Dai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - George F Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
981
|
Datta S, Hett EC, Vora KA, Hazuda DJ, Oslund RC, Fadeyi OO, Emili A. The chemical biology of coronavirus host-cell interactions. RSC Chem Biol 2021; 2:30-46. [PMID: 34458775 PMCID: PMC8340996 DOI: 10.1039/d0cb00197j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/06/2020] [Indexed: 12/25/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the current coronavirus disease 2019 (COVID-19) pandemic that has led to a global economic disruption and collapse. With several ongoing efforts to develop vaccines and treatments for COVID-19, understanding the molecular interaction between the coronavirus, host cells, and the immune system is critical for effective therapeutic interventions. Greater insight into these mechanisms will require the contribution and combination of multiple scientific disciplines including the techniques and strategies that have been successfully deployed by chemical biology to tease apart complex biological pathways. We highlight in this review well-established strategies and methods to study coronavirus-host biophysical interactions and discuss the impact chemical biology will have on understanding these interactions at the molecular level.
Collapse
Affiliation(s)
- Suprama Datta
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine Boston MA USA
| | - Erik C Hett
- Exploratory Science Center, Merck & Co., Inc. Cambridge Massachusetts USA
| | - Kalpit A Vora
- Infectious Diseases and Vaccine Research, Merck & Co., Inc. West Point Pennsylvania USA
| | - Daria J Hazuda
- Exploratory Science Center, Merck & Co., Inc. Cambridge Massachusetts USA
- Infectious Diseases and Vaccine Research, Merck & Co., Inc. West Point Pennsylvania USA
| | - Rob C Oslund
- Exploratory Science Center, Merck & Co., Inc. Cambridge Massachusetts USA
| | | | - Andrew Emili
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine Boston MA USA
| |
Collapse
|
982
|
Das UN. Essential fatty acids and their metabolites in the pathobiology of (coronavirus disease 2019) COVID-19. Nutrition 2021; 82:111052. [PMID: 33290970 PMCID: PMC7657034 DOI: 10.1016/j.nut.2020.111052] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/02/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Undurti N Das
- UND Life Sciences, Battle Ground, WA, USA; BioScience Research Centre and Department of Medicine, GVP Medical College and Hospital, Visakhapatnam, India.
| |
Collapse
|
983
|
Rendeiro AF, Casano J, Vorkas CK, Singh H, Morales A, DeSimone RA, Ellsworth GB, Soave R, Kapadia SN, Saito K, Brown CD, Hsu J, Kyriakides C, Chiu S, Cappelli LV, Cacciapuoti MT, Tam W, Galluzzi L, Simonson PD, Elemento O, Salvatore M, Inghirami G. Profiling of immune dysfunction in COVID-19 patients allows early prediction of disease progression. Life Sci Alliance 2021; 4:e202000955. [PMID: 33361110 PMCID: PMC7768198 DOI: 10.26508/lsa.202000955] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 01/08/2023] Open
Abstract
With a rising incidence of COVID-19-associated morbidity and mortality worldwide, it is critical to elucidate the innate and adaptive immune responses that drive disease severity. We performed longitudinal immune profiling of peripheral blood mononuclear cells from 45 patients and healthy donors. We observed a dynamic immune landscape of innate and adaptive immune cells in disease progression and absolute changes of lymphocyte and myeloid cells in severe versus mild cases or healthy controls. Intubation and death were coupled with selected natural killer cell KIR receptor usage and IgM+ B cells and associated with profound CD4 and CD8 T-cell exhaustion. Pseudo-temporal reconstruction of the hierarchy of disease progression revealed dynamic time changes in the global population recapitulating individual patients and the development of an eight-marker classifier of disease severity. Estimating the effect of clinical progression on the immune response and early assessment of disease progression risks may allow implementation of tailored therapies.
Collapse
Affiliation(s)
- André F Rendeiro
- Institute of Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Joseph Casano
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Charles Kyriakos Vorkas
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Harjot Singh
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ayana Morales
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Robert A DeSimone
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Grant B Ellsworth
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Rosemary Soave
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shashi N Kapadia
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Kohta Saito
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Christopher D Brown
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - JingMei Hsu
- Division of Hematology/Oncology, Department of Medicine Weill Cornell Medicine, New York, NY, USA
| | - Christopher Kyriakides
- Department of Rehabilitation Medicine at New York University Grossman School of Medicine New York, NY, USA
| | - Steven Chiu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Luca Vincenzo Cappelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Wayne Tam
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lorenzo Galluzzi
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Paul D Simonson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Elemento
- Institute of Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Mirella Salvatore
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
- Division of Public Health Programs, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
984
|
Ribas A, Sengupta R, Locke T, Zaidi SK, Campbell KM, Carethers JM, Jaffee EM, Wherry EJ, Soria JC, D'Souza G. Priority COVID-19 Vaccination for Patients with Cancer while Vaccine Supply Is Limited. Cancer Discov 2021; 11:233-236. [PMID: 33355178 PMCID: PMC8053003 DOI: 10.1158/2159-8290.cd-20-1817] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Published series on COVID-19 support the notion that patients with cancer are a particularly vulnerable population. There is a confluence of risk factors between cancer and COVID-19, and cancer care and treatments increase exposure to the virus and may dampen natural immune responses. The available evidence supports the conclusion that patients with cancer, in particular with hematologic malignancies, should be considered among the very high-risk groups for priority COVID-19 vaccination.
Collapse
Affiliation(s)
- Antoni Ribas
- Department of Medicine and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.
| | - Rajarshi Sengupta
- American Association for Cancer Research, Philadelphia, Pennsylvania
| | - Trevan Locke
- American Association for Cancer Research, Philadelphia, Pennsylvania
| | | | - Katie M Campbell
- Department of Medicine and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| | - John M Carethers
- Departments of Internal Medicine and Human Genetics and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Elizabeth M Jaffee
- Department of Medicine, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - E John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jean-Charles Soria
- Paris Saclay University, Saint-Aubin, France and Drug Development Department, Gustave Roussy, Villejuif, France
| | | |
Collapse
|
985
|
Hennessy EJ, FitzGerald GA. Battle for supremacy: nucleic acid interactions between viruses and cells. J Clin Invest 2021; 131:144227. [PMID: 33290272 PMCID: PMC7843224 DOI: 10.1172/jci144227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Since the COVID-19 pandemic swept across the globe, researchers have been trying to understand its origin, life cycle, and pathogenesis. There is a striking variability in the phenotypic response to infection with SARS-CoV-2 that may reflect differences in host genetics and/or immune response. It is known that the human epigenome is influenced by ethnicity, age, lifestyle, and environmental factors, including previous viral infections. This Review examines the influence of viruses on the host epigenome. We describe general lessons and methodologies that can be used to understand how the virus evades the host immune response. We consider how variation in the epigenome may contribute to heterogeneity in the response to SARS-CoV-2 and may identify a precision medicine approach to treatment.
Collapse
|
986
|
Lee P, Kim CU, Seo SH, Kim DJ. Current Status of COVID-19 Vaccine Development: Focusing on Antigen Design and Clinical Trials on Later Stages. Immune Netw 2021; 21:e4. [PMID: 33728097 PMCID: PMC7937514 DOI: 10.4110/in.2021.21.e4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 12/23/2022] Open
Abstract
The global outbreak of coronavirus disease 2019 (COVID-19) is still threatening human health, economy, and social life worldwide. As a counteraction for this devastating disease, a number of vaccines are being developed with unprecedented speed combined with new technologies. As COVID-19 vaccines are being developed in the absence of a licensed human coronavirus vaccine, there remain further questions regarding the long-term efficacy and safety of the vaccines, as well as immunological mechanisms in depth. This review article discusses the current status of COVID-19 vaccine development, mainly focusing on antigen design, clinical trials in later stages, and immunological considerations for further study.
Collapse
Affiliation(s)
- Pureum Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Bioscience, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Chang-Ung Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Korea
| | | | - Doo-Jin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Bioscience, University of Science and Technology (UST), Daejeon 34113, Korea
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
987
|
Najm A, Alunno A, Mariette X, Terrier B, De Marco G, Emmel J, Mason L, McGonagle DG, Machado PM. Pathophysiology of acute respiratory syndrome coronavirus 2 infection: a systematic literature review to inform EULAR points to consider. RMD Open 2021; 7:e001549. [PMID: 33574116 PMCID: PMC7880117 DOI: 10.1136/rmdopen-2020-001549] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/08/2021] [Accepted: 01/14/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The SARS-CoV-2 pandemic is a global health problem. Beside the specific pathogenic effect of SARS-CoV-2, incompletely understood deleterious and aberrant host immune responses play critical roles in severe disease. Our objective was to summarise the available information on the pathophysiology of COVID-19. METHODS Two reviewers independently identified eligible studies according to the following PICO framework: P (population): patients with SARS-CoV-2 infection; I (intervention): any intervention/no intervention; C (comparator): any comparator; O (outcome) any clinical or serological outcome including but not limited to immune cell phenotype and function and serum cytokine concentration. RESULTS Of the 55 496 records yielded, 84 articles were eligible for inclusion according to question-specific research criteria. Proinflammatory cytokine expression, including interleukin-6 (IL-6), was increased, especially in severe COVID-19, although not as high as other states with severe systemic inflammation. The myeloid and lymphoid compartments were differentially affected by SARS-CoV-2 infection depending on disease phenotype. Failure to maintain high interferon (IFN) levels was characteristic of severe forms of COVID-19 and could be related to loss-of-function mutations in the IFN pathway and/or the presence of anti-IFN antibodies. Antibody response to SARS-CoV-2 infection showed a high variability across individuals and disease spectrum. Multiparametric algorithms showed variable diagnostic performances in predicting survival, hospitalisation, disease progression or severity, and mortality. CONCLUSIONS SARS-CoV-2 infection affects both humoral and cellular immunity depending on both disease severity and individual parameters. This systematic literature review informed the EULAR 'points to consider' on COVID-19 pathophysiology and immunomodulatory therapies.
Collapse
Affiliation(s)
- Aurélie Najm
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Alessia Alunno
- Department of Medicine, Rheumatology Unit, University of Perugia, Perugia, Italy
| | - Xavier Mariette
- INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Paris-Sud University, Paris-Saclay University, Le Kremlin-Bicêtre, France
- Department of Rheumatology, AP-HP, Paris-Sud University Hospitals, Le Kremlin Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Benjamin Terrier
- University of Paris, Assistance Publique-Hôpitaux de Paris, Cochin Hospital, Paris, France
- INSERM U970, PARCC, Paris, Île-de-France, France
| | - Gabriele De Marco
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, NIHR Leeds Biomedical Research Centre, Leeds, West Yorkshire, UK
- Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, West Yorkshire, UK
| | - Jenny Emmel
- Medical Education, Library & Evidence Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Laura Mason
- Medical Education, Library & Evidence Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Dennis G McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, NIHR Leeds Biomedical Research Centre, Leeds, West Yorkshire, UK
- Chapel Allerton Hospital, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, NIHR Leeds Biomedical Research Centre, Leeds, UK
| | - Pedro M Machado
- Centre for Rheumatology, National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre (BRC), University College London Hospitals (UCLH) NHS Foundation Trus, London, UK
- Department of Rheumatology, Northwick Park Hospital, London North West University Healthcare NHS Trust, London, UK
- Centre for Rheumatology & Department of Neuromuscular Diseases, University College London, London, UK
| |
Collapse
|
988
|
Hong J, Jhun H, Choi YO, Taitt AS, Bae S, Lee Y, Song CS, Yeom SC, Kim S. Structure of SARS-CoV-2 Spike Glycoprotein for Therapeutic and Preventive Target. Immune Netw 2021; 21:e8. [PMID: 33728101 PMCID: PMC7937506 DOI: 10.4110/in.2021.21.e8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/08/2021] [Accepted: 02/14/2021] [Indexed: 12/14/2022] Open
Abstract
The global crisis caused by the coronavirus disease 2019 (COVID-19) led to the most significant economic loss and human deaths after World War II. The pathogen causing this disease is a novel virus called the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). As of December 2020, there have been 80.2 million confirmed patients, and the mortality rate is known as 2.16% globally. A strategy to protect a host from SARS-CoV-2 is by suppressing intracellular viral replication or preventing viral entry. We focused on the spike glycoprotein that is responsible for the entry of SARS-CoV-2 into the host cell. Recently, the US Food and Drug Administration/EU Medicines Agency authorized a vaccine and antibody to treat COVID-19 patients by emergency use approval in the absence of long-term clinical trials. Both commercial and academic efforts to develop preventive and therapeutic agents continue all over the world. In this review, we present a perspective on current reports about the spike glycoprotein of SARS-CoV-2 as a therapeutic target.
Collapse
Affiliation(s)
- Jaewoo Hong
- Department of Physiology, Daegu Catholic University School of Medicine, Daegu 42472, Korea
- Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD 21702, USA
| | - Hyunjhung Jhun
- Technical Assistance Center, Korea Food Research Institute, Wanju 55365, Korea
| | - Yeo-Ok Choi
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Afeisha S. Taitt
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Suyoung Bae
- Department of Bioequivalence Division for Drug Evaluation, Ministry of Food and Drug Safety, Cheongju 28159, Korea
| | - Youngmin Lee
- Department of Medicine, Pusan Paik Hospital, College of Medicine, Inje University, Busan 47392, Korea
| | - Chang-seon Song
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Su Cheong Yeom
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang 25354, Korea
| | - Soohyun Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- Veterinary Science Research Institute, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
989
|
Byazrova M, Yusubalieva G, Spiridonova A, Efimov G, Mazurov D, Baranov K, Baklaushev V, Filatov A. Pattern of circulating SARS-CoV-2-specific antibody-secreting and memory B-cell generation in patients with acute COVID-19. Clin Transl Immunology 2021; 10:e1245. [PMID: 33552508 PMCID: PMC7848539 DOI: 10.1002/cti2.1245] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES To predict the spread of coronavirus disease (COVID-19), information regarding the immunological memory for disease-specific antigens is necessary. The possibility of reinfection, as well as the efficacy of vaccines for COVID-19 that are currently under development, will largely depend on the quality and longevity of immunological memory in patients. To elucidate the process of humoral immunity development, we analysed the generation of plasmablasts and virus receptor-binding domain (RBD)-specific memory B (Bmem) cells in patients during the acute phase of COVID-19. METHODS The frequencies of RBD-binding plasmablasts and RBD-specific antibody-secreting cells (ASCs) in the peripheral blood samples collected from patients with COVID-19 were measured using flow cytometry and the ELISpot assay. RESULTS The acute phase of COVID-19 was characterised by the transient appearance of total as well as RBD-binding plasmablasts. ELISpot analysis indicated that most patients exhibited a spontaneous secretion of RBD-specific ASCs in the circulation with good correlation between the IgG and IgM subsets. IL-21/CD40L stimulation of purified B cells induced the activation and proliferation of Bmem cells, which led to the generation of plasmablast phenotypic cells as well as RBD-specific ASCs. No correlation was observed between the frequency of Bmem cell-derived and spontaneous ASCs, suggesting that the two types of ASCs were weakly associated with each other. CONCLUSION Our findings reveal that SARS-CoV-2-specific Bmem cells are generated during the acute phase of COVID-19. These findings can serve as a basis for further studies on the longevity of SARS-CoV-2-specific B-cell memory.
Collapse
Affiliation(s)
- Maria Byazrova
- National Research Center Institute of Immunology of Federal Medical Biological Agency of RussiaMoscowRussia
- Department of ImmunologyFaculty of BiologyLomonosov Moscow State UniversityMoscowRussia
| | - Gaukhar Yusubalieva
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies of the FMBA of RussiaMoscowRussia
| | - Anna Spiridonova
- National Research Center Institute of Immunology of Federal Medical Biological Agency of RussiaMoscowRussia
| | | | - Dmitriy Mazurov
- Institute of Gene Biology Russian Academy of SciencesCenter for Precision Genome Editing and Genetic Technologies for BiomedicineMoscowRussia
| | - Konstantin Baranov
- Institute of Molecular and Cellular Biology SB RASLomonosov Moscow State UniversityNovosibirskRussia
| | - Vladimir Baklaushev
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies of the FMBA of RussiaMoscowRussia
| | - Alexander Filatov
- National Research Center Institute of Immunology of Federal Medical Biological Agency of RussiaMoscowRussia
- Department of ImmunologyFaculty of BiologyLomonosov Moscow State UniversityMoscowRussia
| |
Collapse
|
990
|
DiPiazza AT, Graham BS, Ruckwardt TJ. T cell immunity to SARS-CoV-2 following natural infection and vaccination. Biochem Biophys Res Commun 2021; 538:211-217. [PMID: 33190827 PMCID: PMC7584424 DOI: 10.1016/j.bbrc.2020.10.060] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022]
Abstract
SARS-CoV-2 first emerged in the human population in late 2019 in Wuhan, China, and in a matter of months, spread across the globe resulting in the Coronavirus Disease 19 (COVID-19) pandemic and substantial economic fallout. SARS-CoV-2 is transmitted between humans via respiratory particles, with infection presenting a spectrum of clinical manifestations ranging from asymptomatic to respiratory failure with multiorgan dysfunction and death in severe cases. Prior experiences with human pathogenic coronaviruses and respiratory virus diseases in general have revealed an important role for cellular immunity in limiting disease severity. Here, we review some of the key mechanisms underlying cell-mediated immunity to respiratory viruses and summarize our current understanding of the functional capacity and role of SARS-CoV-2-specific T cells following natural infection and vaccination.
Collapse
Affiliation(s)
- Anthony T DiPiazza
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, USA.
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Tracy J Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, USA.
| |
Collapse
|
991
|
Lost in deletion: The enigmatic ORF8 protein of SARS-CoV-2. Biochem Biophys Res Commun 2021; 538:116-124. [PMID: 33685621 PMCID: PMC7577707 DOI: 10.1016/j.bbrc.2020.10.045] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/18/2020] [Indexed: 12/15/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome contains nine open reading frames (ORFs) that encode for accessory proteins which, although dispensable for viral replication, are important for the modulation of the host infected cell metabolism and innate immunity evasion. Among those, the ORF8 gene encodes for the homonymous multifunctional, highly immunogenic, immunoglobulin-like protein that was recently found to inhibit presentation of viral antigens by class I major histocompatibility complex, suppress the type I interferon antiviral response and interact with host factors involved in pulmonary inflammation and fibrogenesis. Moreover, the ORF8 is a hypervariable gene rapidly evolving among SARS-related coronaviruses, with a tendency to recombine and undergo deletions that are deemed to facilitate the virus adaptation to the human host. Intriguingly, SARS-CoV-2 variants isolated in the beginning of the coronavirus disease 2019 (Covid-19) pandemic that were deleted of the ORF8 gene have been associated to milder symptoms and better disease outcome. This minireview summarizes the current knowledge on the SARS-CoV-2 ORF8 protein in perspective to its potential as antiviral target and with special emphasis on the biochemical, biophysical and structural aspects of its molecular biology.
Collapse
|
992
|
Kenny G, Mallon PW. COVID19- clinical presentation and therapeutic considerations. Biochem Biophys Res Commun 2021; 538:125-131. [PMID: 33218685 PMCID: PMC7657079 DOI: 10.1016/j.bbrc.2020.11.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 11/07/2020] [Indexed: 12/15/2022]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as a pandemic infection in 2020 has presented many therapeutic challenges. Not least among these is the importance of abnormal host response to infection that is one of the main drivers of more severe disease. Despite significant research endeavours, very few effective therapies have been identified, in part related to the different pathogenic mechanisms underlying different stages of clinical COVID-19. This mini review summarises data related to current and potential future therapies for COVID-19 and highlights the many challenges inherent in developing effective therapeutic options for new pandemic infection.
Collapse
Affiliation(s)
- Grace Kenny
- Centre for Experimental Pathogen Host Research, University College Dublin, Ireland; Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland.
| | - Patrick W Mallon
- Centre for Experimental Pathogen Host Research, University College Dublin, Ireland; Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
993
|
Fraga-Silva TFDC, Maruyama SR, Sorgi CA, Russo EMDS, Fernandes APM, de Barros Cardoso CR, Faccioli LH, Dias-Baruffi M, Bonato VLD. COVID-19: Integrating the Complexity of Systemic and Pulmonary Immunopathology to Identify Biomarkers for Different Outcomes. Front Immunol 2021; 11:599736. [PMID: 33584667 PMCID: PMC7878380 DOI: 10.3389/fimmu.2020.599736] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
In the last few months, the coronavirus disease 2019 (COVID-19) pandemic has affected millions of people worldwide and has provoked an exceptional effort from the scientific community to understand the disease. Clinical evidence suggests that severe COVID-19 is associated with both dysregulation of damage tolerance caused by pulmonary immunopathology and high viral load. In this review article, we describe and discuss clinical studies that show advances in the understanding of mild and severe illness and we highlight major points that are critical for improving the comprehension of different clinical outcomes. The understanding of pulmonary immunopathology will contribute to the identification of biomarkers in an attempt to classify mild, moderate, severe and critical COVID-19 illness. The interface of pulmonary immunopathology and the identification of biomarkers are critical for the development of new therapeutic strategies aimed to reduce the systemic and pulmonary hyperinflammation in severe COVID-19.
Collapse
Affiliation(s)
- Thais Fernanda de Campos Fraga-Silva
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Sandra Regina Maruyama
- Department of Genetics and Evolution, Federal University of Sao Carlos, Sao Carlos, Brazil
| | - Carlos Arterio Sorgi
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Elisa Maria de Sousa Russo
- Department of Clinical Analysis, Toxicological and Food Science Analysis, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Ana Paula Morais Fernandes
- Department of General and Specialized Nursing, School of Nursing of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Cristina Ribeiro de Barros Cardoso
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
- Department of Clinical Analysis, Toxicological and Food Science Analysis, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Lucia Helena Faccioli
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
- Department of Clinical Analysis, Toxicological and Food Science Analysis, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Marcelo Dias-Baruffi
- Department of Clinical Analysis, Toxicological and Food Science Analysis, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Vânia Luiza Deperon Bonato
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
994
|
Switched and unswitched memory B cells detected during SARS-CoV-2 convalescence correlate with limited symptom duration. PLoS One 2021; 16:e0244855. [PMID: 33507994 PMCID: PMC7843013 DOI: 10.1371/journal.pone.0244855] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/18/2020] [Indexed: 12/17/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of the pandemic human respiratory illness COVID-19, is a global health emergency. While severe acute disease has been linked to an expansion of antibody-secreting plasmablasts, we sought to identify B cell responses that correlated with positive clinical outcomes in convalescent patients. We characterized the peripheral blood B cell immunophenotype and plasma antibody responses in 40 recovered non-hospitalized COVID-19 subjects that were enrolled as donors in a convalescent plasma treatment study. We observed a significant negative correlation between the frequency of peripheral blood memory B cells and the duration of symptoms for convalescent subjects. Memory B cell subsets in convalescent subjects were composed of classical CD24+ class-switched memory B cells, but also activated CD24-negative and natural unswitched CD27+ IgD+ IgM+ subsets. Memory B cell frequency was significantly correlated with both IgG1 and IgM responses to the SARS-CoV-2 spike protein receptor binding domain (RBD) in most seropositive subjects. IgM+ memory, but not switched memory, directly correlated with virus-specific antibody responses, and remained stable over 3 months. Our findings suggest that the frequency of memory B cells is a critical indicator of disease resolution, and that IgM+ memory B cells may play an important role in SARS-CoV-2 immunity.
Collapse
|
995
|
The immune roadmap for understanding multi-system inflammatory syndrome in children: opportunities and challenges. Nat Med 2021; 26:1819-1824. [PMID: 33139949 DOI: 10.1038/s41591-020-1140-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
996
|
Wijeratne T, Gillard Crewther S, Sales C, Karimi L. COVID-19 Pathophysiology Predicts That Ischemic Stroke Occurrence Is an Expectation, Not an Exception-A Systematic Review. Front Neurol 2021; 11:607221. [PMID: 33584506 PMCID: PMC7876298 DOI: 10.3389/fneur.2020.607221] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/30/2020] [Indexed: 12/27/2022] Open
Abstract
Clinical reports of neurological manifestations associated with severe coronavirus disease 2019 (COVID-19), such as acute ischemic stroke (AIS), encephalopathy, seizures, headaches, acute necrotizing encephalitis, cerebral microbleeds, posterior reversible leukoencephalopathy syndrome, hemophagocytic lymphohistiocytosis, peripheral neuropathy, cranial nerve palsies, transverse myelitis, and demyelinating disorders, are increasing rapidly. However, there are comparatively few studies investigating the potential impact of immunological responses secondary to hypoxia, oxidative stress, and excessive platelet-induced aggregation on the brain. This scoping review has focused on the pathophysiological mechanisms associated with peripheral and consequential neural (central) inflammation leading to COVID-19-related ischemic strokes. It also highlights the common biological processes shared between AIS and COVID-19 infection and the importance of the recognition that severe respiratory dysfunction and neurological impairments associated with COVID and chronic inflammation [post-COVID-19 neurological syndrome (PCNS)] may significantly impact recovery and ability to benefit from neurorehabilitation. This study provides a comprehensive review of the pathobiology of COVID-19 and ischemic stroke. It also affirms that the immunological contribution to the pathophysiology of COVID-19 is predictive of the neurological sequelae particularly ischemic stroke, which makes it the expectation rather than the exception. This work is of fundamental significance to the neurorehabilitation community given the increasing number of COVID-related ischemic strokes, the current limited knowledge regarding the risk of reinfection, and recent reports of a PCNS. It further highlights the need for global collaboration and research into new pathobiology-based neurorehabilitation treatment strategies and more integrated evidence-based care.
Collapse
Affiliation(s)
- Tissa Wijeratne
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
- Department of Neurology, Western Health and University Melbourne, Australian Institute of Muscular Skeletal Sciences (AIMSS), Level Three, Western Health Centre for Research and Education (WHCRE), Sunshine Hospital, Melbourne, VIC, Australia
- Department of Medicine, Faculty of Medicine, University of Rajarata, Anuradhapura, Sri Lanka
| | - Sheila Gillard Crewther
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
- Department of Neurology, Western Health and University Melbourne, Australian Institute of Muscular Skeletal Sciences (AIMSS), Level Three, Western Health Centre for Research and Education (WHCRE), Sunshine Hospital, Melbourne, VIC, Australia
| | - Carmela Sales
- Department of Neurology, Western Health and University Melbourne, Australian Institute of Muscular Skeletal Sciences (AIMSS), Level Three, Western Health Centre for Research and Education (WHCRE), Sunshine Hospital, Melbourne, VIC, Australia
- Department of Medicine, Faculty of Medicine, University of Rajarata, Anuradhapura, Sri Lanka
| | - Leila Karimi
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
- Department of Neurology, Western Health and University Melbourne, Australian Institute of Muscular Skeletal Sciences (AIMSS), Level Three, Western Health Centre for Research and Education (WHCRE), Sunshine Hospital, Melbourne, VIC, Australia
- Faculty of Social and Political Sciences, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| |
Collapse
|
997
|
Affiliation(s)
- Erin K McCreary
- Department of Medicine, Division of Infectious Diseases, UPMC Health System and the University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Nuala J Meyer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
998
|
Affiliation(s)
- Robin Cherian
- Department of Cardiology, National University Heart Centre Singapore, Singapore
| |
Collapse
|
999
|
Rezaei M, Marjani M, Mahmoudi S, Mortaz E, Mansouri D. Dynamic Changes of Lymphocyte Subsets in the Course of COVID-19. Int Arch Allergy Immunol 2021; 182:254-262. [PMID: 33498051 PMCID: PMC7900469 DOI: 10.1159/000514202] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Background Although the pathophysiology of coronavirus disease 2019 (COVID-19) is not clearly defined, among the proposed mechanisms, immune system dysfunction is more likely than others. The aim of this study was to clarify the characteristics and clinical significance of dynamic changes of lymphocyte subsets in the course of COVID-19. Methods In this prospective study, the levels of peripheral lymphocyte subsets including CD4+, CD8+, CD4+CD25+FOXP3+, CD38+, CD3+HLA-DR+, CD19+, CD20+, and CD16+CD56+ cells were measured by flow cytometry in 52 confirmed hospitalized patients with COVID-19 at the day of admission and after 7 days of care. Clinical response was defined as improvement in symptoms (fever, dyspnea, and cough as well as blood oxygen saturation), and patients who met these criteria after 1 week of admission were classified as early responders; others who survived and finally discharged from the hospital were classified as late responders and patients who died were categorized as nonresponders. Immunophenotyping of studied cell changes on the first day of admission and 7 days after treatment were compared. Besides, the correlation between cellular subset variation and clinical response and outcome were analyzed. Results Total counts of white blood cell, T cells, CD4+ T cells, CD8+ T cells, CD38+ lymphocytes, and CD3+HLA-DR+ lymphocytes were significantly increased in both early and late responders. No statistically significant difference was observed in CD4+/CD8+ ratio, B cells, FOXP3+T<sub>reg</sub> lymphocytes, and FOXP3 median fluorescence intensity among studied groups. According to the multivariate analysis, an increase in CD4+ T cells (p = 0.019), CD8+ T cells (p = 0.001), and administration of interferon (p < 0.001) were independent predictors of clinical response. Conclusion We found an increasing trend in total T cells, T helpers, cytotoxic T cells, activated lymphocytes, and natural killer cells among responders. This trend was not statistically significant among nonresponders. The findings of this study may enhance our knowledge about the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Mitra Rezaei
- Virology Research Center, National Research Institute of Tuberculosis and Lung diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Marjani
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran,
| | - Shima Mahmoudi
- Pediatric Infectious Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Mortaz
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Mansouri
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
1000
|
Liu L, Chen Z, Du Y, Gao J, Li J, Deng T, Chen C, Wang L, Yang Y, Liu C. CD8 + T cells predicted the conversion of common covid-19 to severe. Sci Rep 2021; 11:2169. [PMID: 33500507 PMCID: PMC7838185 DOI: 10.1038/s41598-021-81732-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 01/11/2021] [Indexed: 01/13/2023] Open
Abstract
To evaluate the predictive effect of T-lymphoid subsets on the conversion of common covid-19 to severe. The laboratory data were collected retrospectively from common covid-19 patients in the First People's Hospital of Zaoyang, Hubei Province, China and the Third People's Hospital of Kunming, Yunnan Province, China, between January 20, 2020 and March 15, 2020 and divided into training set and validation set. Univariate and multivariate logistic regression was performed to investigate the risk factors for the conversion of common covid-19 to severe in the training set, the prediction model was established and verified externally in the validation set. 60 (14.71%) of 408 patients with common covid-19 became severe in 6-10 days after diagnosis. Univariate and multiple logistic regression analysis revealed that lactate (P = 0.042, OR = 1097.983, 95% CI 1.303, 924,798.262) and CD8+ T cells (P = 0.010, OR = 0.903, 95% CI 0.835, 0.975) were independent risk factors for general type patients to turn to severe type. The area under ROC curve of lactate and CD8+ T cells was 0.754 (0.581, 0.928) and 0.842 (0.713, 0.970), respectively. The actual observation value was highly consistent with the prediction model value in curve fitting. The established prediction model was verified in 78 COVID-19 patients in the verification set, the area under the ROC curve was 0.906 (0.861, 0.981), and the calibration curve was consistent. CD8+ T cells, as an independent risk factor, could predict the transition from common covid-19 to severe.
Collapse
Affiliation(s)
- Li Liu
- Department of Liver Disease, The Third People's Hospital of Kunming City, Yunnan, China.
| | - Zhiyong Chen
- Department of Infectious Diseases, The First People's Hospital of Zaoyang City, Hubei, China
| | - Yingrong Du
- Department of Liver Disease, The Third People's Hospital of Kunming City, Yunnan, China
| | - Jianpeng Gao
- Department of Liver Disease, The Third People's Hospital of Kunming City, Yunnan, China
| | - Junyi Li
- Department of Liver Disease, The Third People's Hospital of Kunming City, Yunnan, China
| | - Tiqin Deng
- Department of Infectious Diseases, The First People's Hospital of Zaoyang City, Hubei, China
| | - Chen Chen
- Department of Infectious Diseases, The First People's Hospital of Zaoyang City, Hubei, China
| | - Lin Wang
- Department of Liver Disease, The Third People's Hospital of Kunming City, Yunnan, China
| | - Yongrui Yang
- Department of Liver Disease, The Third People's Hospital of Kunming City, Yunnan, China
| | - Chunyun Liu
- Department of Liver Disease, The Third People's Hospital of Kunming City, Yunnan, China
| |
Collapse
|