1
|
Rovito R, Bono V, Coianiz N, Cazzetta V, Franzese S, Mikulak J, Di Vito C, Bai F, Beaudoin-Bussières G, Tauzin A, Augello M, Tincati C, Santoro A, Borghi E, Marozin S, Finzi A, Della Bella S, Mavilio D, Marchetti G. Multi-layered deep immune profiling, SARS-CoV-2 RNAemia and inflammation in unvaccinated COVID-19 individuals with persistent symptoms. COMMUNICATIONS MEDICINE 2025; 5:155. [PMID: 40325175 PMCID: PMC12052991 DOI: 10.1038/s43856-025-00832-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/28/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Long-COVID immunopathogenesis involves diverse factors. We longitudinally characterize hospitalized COVID-19 patients, examining the role of SARS-CoV-2 RNAemia and inflammation in immune dysregulation. METHODS Hospitalized patients are evaluated during acute infection (T0), 3 months post-symptom onset (T1), and 3 years if symptoms persisted (T2). Immune profile includes characterization of SARS-CoV-2-specific/non-specific T/B cells (flow cytometry) and antibodies (ELISA, neutralization, ADCC). RNAemia and cytokines are quantified (RT-PCR, cytometric beads array) and correlated. STATISTICS non-parametric cross-sectional, longitudinal and correlation analyses. RESULTS Here we show 48 hospitalized individuals during acute COVID-19, 38 exhibit early persistent symptoms (EPS+) 3 months post-symptoms onset, 10 do not (EPS-). Groups are comparable for age, sex, co-morbidities. The EPS+ shows fatigue, dyspnoea, anosmia/dysgeusia, diarrhea, chronic pain, mnestic disorders. Over time, they show a reduction of neutralization ability and total SARS-CoV-2-specific CD4 T cells, with increased total CD4 TEMRA, and failure to increase RBD-specific B cells and IgA+ MBCs. EPS+ patients show higher levels of T0-IFN-γ + CD4 TEMRA, T1-IL-2 + CD4 TEM and T1-TNF-α + CD4 cTfh. In EPS+, baseline SARS-CoV-2 RNAemia positively correlates with CD4 TEMRA, follow-up SARS-CoV-2 RNAemia with ADCC. Among 38 EPS+ individuals at T1, 33 are evaluated 3 years after infection, 5 are lost at follow-up. 10/33 EPS+ show long-term symptoms (late persistent symptoms, EPS + LPS+), whereas 23/33 fully recover (EPS + LPS-). Antibodies, RNAemia, and cytokines show no differences between/within groups at any time point. CONCLUSIONS Early persistent symptoms are associated with multi-layered SARS-CoV-2-specific/non-SARS-CoV-2-specific immune dysregulation. The shift towards non-Ag-specific TEMRA and ADCC trigger in EPS+ may relate to SARS-CoV-2 RNAemia. Early immune dysregulation does not associate with long-term persistent symptoms. Further research on SARS-CoV-2 RNAemia and early immune dysregulation is needed.
Collapse
Affiliation(s)
- Roberta Rovito
- Clinic of Infectious Diseases and Tropical Medicine, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Valeria Bono
- Clinic of Infectious Diseases and Tropical Medicine, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Nicolò Coianiz
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Innate Lymphoid Cells and Cancer, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Valentina Cazzetta
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sara Franzese
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Joanna Mikulak
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Clara Di Vito
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Francesca Bai
- Clinic of Infectious Diseases and Tropical Medicine, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Guillaume Beaudoin-Bussières
- Centre de recherche du CHUM (CRCHUM), Montréal, QC, H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Alexandra Tauzin
- Centre de recherche du CHUM (CRCHUM), Montréal, QC, H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Matteo Augello
- Clinic of Infectious Diseases and Tropical Medicine, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Camilla Tincati
- Clinic of Infectious Diseases and Tropical Medicine, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Andrea Santoro
- Clinic of Infectious Diseases and Tropical Medicine, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Elisa Borghi
- Clinical Microbiology, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Sabrina Marozin
- Clinic of Infectious Diseases and Tropical Medicine, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Andrés Finzi
- Centre de recherche du CHUM (CRCHUM), Montréal, QC, H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Silvia Della Bella
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Domenico Mavilio
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Giulia Marchetti
- Clinic of Infectious Diseases and Tropical Medicine, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy.
| |
Collapse
|
2
|
Gong P, Lu Y, Chai X, Li X. Exploring the Causal Relationship Between Immune Cells and Idiopathic Pulmonary Fibrosis: A Mendelian Randomization Analysis. J Clin Lab Anal 2025; 39:e70026. [PMID: 40167279 PMCID: PMC12019702 DOI: 10.1002/jcla.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible interstitial lung disease with a complex pathogenesis involving multiple immune cells. This study investigates the relationship between immune cells and IPF using Mendelian randomization (MR) analysis. METHODS A two-sample MR analysis was performed using genome-wide association studies (GWAS) and immune cell databases by R software. We analyzed data from 1028 European individuals with IPF, focusing on 731 immune traits. The primary method of analysis was inverse variance weighting (IVW), supplemented with sensitivity analyses, including MR-Egger regression and MR-PRESSO, to detect and correct for pleiotropy. RESULTS The MR analysis identified six immune panels and 23 immune traits significantly associated with IPF, including five traits that increase and 18 traits that decrease IPF risk. Notable traits increasing IPF risk included switched memory B-cells (OR = 1.27, p = 0.0029) and IgD- CD38dim B-cells (OR = 1.08, p = 0.0449). Traits associated with a reduced IPF risk included central memory CD4+ T-cells (%CD4+, OR = 0.96, p = 0.0489), CD20 on naive-mature B-cells (OR = 0.94, p = 0.0499), and CD33br HLA-DR+ absolute count (AC) (OR = 0.93, p = 0.0489). There was no significant causal relationship between IPF disease and some immune traits (p > 0.05). CONCLUSION This study suggests a potential causal link between specific immune cell traits and the development of IPF, providing new insights into the disease's immunological mechanisms. Future research should focus on validating these findings in larger, more diverse populations to inform drug development and therapeutic strategies.
Collapse
Affiliation(s)
- Peng Gong
- School of Basic Medical SciencesShanxi University of Traditional Chinese MedicineJinzhongShanxiChina
- Shouyang County People's HospitalShouyangShanxiChina
| | - Yimin Lu
- School of Basic Medical SciencesShanxi University of Traditional Chinese MedicineJinzhongShanxiChina
| | - Xi Chai
- School of Basic Medical SciencesShanxi University of Traditional Chinese MedicineJinzhongShanxiChina
| | - Xiaobo Li
- School of Basic Medical SciencesShanxi University of Traditional Chinese MedicineJinzhongShanxiChina
| |
Collapse
|
3
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
4
|
Flores-Gonzalez J, Buendia-Roldan I, Téllez-Quijada F, Peña-Bates C, Ramón-Luing LA, Castorena-Maldonado A, Falfán-Valencia R, Pérez-Rubio G, Selman M, Chavez-Galan L, Chávez-Galán L. Altered immune surveillance of B and T cells in patients with persistent residual lung abnormalities 12 months after severe COVID-19. Respir Res 2025; 26:22. [PMID: 39827348 PMCID: PMC11742501 DOI: 10.1186/s12931-025-03102-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/05/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Post-COVID-19 respiratory sequelae often involve lung damage, which is called residual lung abnormalities, and potentially lead to chronic respiratory issues. The adaptive immune response, involving T-cells and B-cells, plays a critical role in pathogen control, inflammation, and tissue repair. However, the link between immune dysregulation and the development of residual lung abnormalities remains unclear. METHODS 109 patients discharged with residual lung abnormalities after a critical COVID-19 were followed for 12 months and divided as full recovery patients (FRG, n = 88) and persistent lung abnormalities (PLAG, n = 21). Cell profiling analysis was done using flow cytometry at 24 h of not antigen-specific in vitro stimulation. Plasma or supernatant levels of IFN-g, IL-4, IL-10, IgM, and IgG were assessed, and 10 patients (5 FRG, 5 PLAG) were randomly selected for detailed immune cell phenotyping and functional analysis of peripheral blood mononuclear cells using flow cytometry. RESULTS Compared to the FRG group, PLAG exhibited an increase of unswitched (p = 0.0159) and decreased double-negative activated B-cells (p = 0.0317), systemic IL-10 levels were lower, displayed reduced frequency of total B-cells, and impaired spontaneous IgM (p = 0.0357) and IgG (p = 0.0079) release in culture. Regarding T-cells, PLAG patients showed a reduction in effector memory CD4 + cells (p = 0.0159) and an increase in CD4 + TEMRA cells (p = 0.0079) following in vitro stimulation. Notably, the PLAG group also exhibited higher frequencies of central memory CD4 + Th2 (GATA3+) T-cells in response to activation than the FRG group (p = 0.0079). CONCLUSIONS Patients with residual lung abnormalities 12 months post-critical COVID-19 exhibit impaired B-cell function, increased unswitched B-cells, and higher frequencies of CD4 + TEMRA T-cells following in vitro activation. These immune imbalances may contribute to ongoing lung dysfunction and warrant further investigation as a potential mechanism in residual lung abnormalities. Larger studies are necessary to confirm these findings.
Collapse
Affiliation(s)
- Julio Flores-Gonzalez
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Ivette Buendia-Roldan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Fernanda Téllez-Quijada
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Carlos Peña-Bates
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Lucero A Ramón-Luing
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | | | - Ramcés Falfán-Valencia
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Gloria Pérez-Rubio
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Leslie Chavez-Galan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico.
| | - Leslie Chávez-Galán
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| |
Collapse
|
5
|
Flores-González J, Monroy-Rodríguez Z, Falfán-Valencia R, Buendía-Roldán I, Fricke-Galindo I, Hernández-Zenteno R, Herrera-Sicairos R, Chávez-Galán L, Pérez-Rubio G. Variants rs3804099 and rs3804100 in the TLR2 Gene Induce Different Profiles of TLR-2 Expression and Cytokines in Response to Spike of SARS-CoV-2. Int J Mol Sci 2024; 25:11063. [PMID: 39456843 PMCID: PMC11507191 DOI: 10.3390/ijms252011063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
The present study aimed to identify in patients with severe COVID-19 and acute respiratory distress syndrome (ARDS) the association between rs3804099 and rs3804100 (TLR2) and evaluate the expression of TLR-2 on the cell surface of innate and adaptive cells of patients' carriers of C allele in at least one genetic variant. We genotyped 1018 patients with COVID-19 and ARDS. According to genotype, a subgroup of 12 patients was selected to stimulate peripheral blood mononuclear cells (PBMCs) with spike and LPS + spike. We evaluated soluble molecules in cell culture supernatants. The C allele in TLR2 (rs3804099, rs3804100) is not associated with a risk of severe COVID-19; however, the presence of the C allele (rs3804099 or rs3804100) affects the TLR-2 ability to respond to a spike of SARS-CoV-2 correctly. The reference group (genotype TT) downregulated the frequency of non-switched TLR-2+ B cells in response to spike stimulus; however, the allele's C carriers group is unable to induce this regulation, but they produce high levels of IL-10, IL-6, and TNF-α by an independent pathway of TLR-2. Findings showed that TT genotypes (rs3804099 and rs3804100) affect the non-switched TLR-2+ B cell distribution. Genotype TT (rs3804099 and rs3804100) affects the TLR-2's ability to respond to a spike of SARS-CoV-2. However, the C allele had increased IL-10, IL-6, and TNF-α by stimulation with spike and LPS.
Collapse
Affiliation(s)
- Julio Flores-González
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (J.F.-G.); (R.H.-S.)
| | - Zurisadai Monroy-Rodríguez
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (Z.M.-R.); (R.F.-V.); (I.F.-G.)
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (Z.M.-R.); (R.F.-V.); (I.F.-G.)
| | - Ivette Buendía-Roldán
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Ingrid Fricke-Galindo
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (Z.M.-R.); (R.F.-V.); (I.F.-G.)
| | - Rafael Hernández-Zenteno
- COPD Clinic, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Ricardo Herrera-Sicairos
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (J.F.-G.); (R.H.-S.)
| | - Leslie Chávez-Galán
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (J.F.-G.); (R.H.-S.)
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (Z.M.-R.); (R.F.-V.); (I.F.-G.)
| |
Collapse
|
6
|
Noviello M, De Lorenzo R, Chimienti R, Maugeri N, De Lalla C, Siracusano G, Lorè NI, Rancoita PMV, Cugnata F, Tassi E, Dispinseri S, Abbati D, Beretta V, Ruggiero E, Manfredi F, Merolla A, Cantarelli E, Tresoldi C, Pastori C, Caccia R, Sironi F, Marzinotto I, Saliu F, Ghezzi S, Lampasona V, Vicenzi E, Cinque P, Manfredi AA, Scarlatti G, Dellabona P, Lopalco L, Di Serio C, Malnati M, Ciceri F, Rovere-Querini P, Bonini C. The longitudinal characterization of immune responses in COVID-19 patients reveals novel prognostic signatures for disease severity, patients' survival and long COVID. Front Immunol 2024; 15:1381091. [PMID: 39136010 PMCID: PMC11317765 DOI: 10.3389/fimmu.2024.1381091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/07/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction SARS-CoV-2 pandemic still poses a significant burden on global health and economy, especially for symptoms persisting beyond the acute disease. COVID-19 manifests with various degrees of severity and the identification of early biomarkers capable of stratifying patient based on risk of progression could allow tailored treatments. Methods We longitudinally analyzed 67 patients, classified according to a WHO ordinal scale as having Mild, Moderate, or Severe COVID-19. Peripheral blood samples were prospectively collected at hospital admission and during a 6-month follow-up after discharge. Several subsets and markers of the innate and adaptive immunity were monitored as putative factors associated with COVID-19 symptoms. Results More than 50 immunological parameters were associated with disease severity. A decision tree including the main clinical, laboratory, and biological variables at admission identified low NK-cell precursors and CD14+CD91+ monocytes, and high CD8+ Effector Memory T cell frequencies as the most robust immunological correlates of COVID-19 severity and reduced survival. Moreover, low regulatory B-cell frequency at one month was associated with the susceptibility to develop long COVID at six months, likely due to their immunomodulatory ability. Discussion These results highlight the profound perturbation of the immune response during COVID-19. The evaluation of specific innate and adaptive immune-cell subsets allows to distinguish between different acute and persistent COVID-19 symptoms.
Collapse
Affiliation(s)
- Maddalena Noviello
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Rebecca De Lorenzo
- Vita-Salute San Raffaele University, Milan, Italy
- Internal Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Raniero Chimienti
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Norma Maugeri
- Autoimmunity and Vascular Inflammation Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Claudia De Lalla
- Experimental Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Gabriel Siracusano
- Immunobiology of Human Immunodeficiency Virus (HIV) Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Nicola Ivan Lorè
- Emerging Bacterial Pathogens Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Paola Maria Vittoria Rancoita
- University Centre for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Federica Cugnata
- University Centre for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Tassi
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Stefania Dispinseri
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Danilo Abbati
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Beretta
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Manfredi
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Aurora Merolla
- Vita-Salute San Raffaele University, Milan, Italy
- Internal Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Cantarelli
- Biological Resource Center Centro Risorse Biologiche-Ospedale San Raffaele (CRB-OSR), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Tresoldi
- Biological Resource Center Centro Risorse Biologiche-Ospedale San Raffaele (CRB-OSR), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Pastori
- Immunobiology of Human Immunodeficiency Virus (HIV) Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Caccia
- Neurovirology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sironi
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Marzinotto
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Fabio Saliu
- Emerging Bacterial Pathogens Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Ghezzi
- Viral Pathogenesis and Biosafety Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Vito Lampasona
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Elisa Vicenzi
- Viral Pathogenesis and Biosafety Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Paola Cinque
- Neurovirology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Angelo Andrea Manfredi
- Autoimmunity and Vascular Inflammation Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Dellabona
- Experimental Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Lopalco
- Immunobiology of Human Immunodeficiency Virus (HIV) Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Clelia Di Serio
- University Centre for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Mauro Malnati
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy
- Hematology and Bone Marrow Transplant Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Patrizia Rovere-Querini
- Vita-Salute San Raffaele University, Milan, Italy
- Internal Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
7
|
Berger L, Wolf J, Kalbitz S, Kellner N, Lübbert C, Borte S. Comparative Analysis of Lymphocyte Populations in Post-COVID-19 Condition and COVID-19 Convalescent Individuals. Diagnostics (Basel) 2024; 14:1286. [PMID: 38928701 PMCID: PMC11202600 DOI: 10.3390/diagnostics14121286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Reduced lymphocyte counts in peripheral blood are one of the most common observations in acute phases of viral infections. Although many studies have already examined the impact of immune (dys)regulation during SARS-CoV-2 infection, there are still uncertainties about the long-term consequences for lymphocyte homeostasis. Furthermore, as persistent cellular aberrations have been described following other viral infections, patients with "Post-COVID-19 Condition" (PCC) may present similarly. In order to investigate cellular changes in the adaptive immune system, we performed a retrospective analysis of flow cytometric data from lymphocyte subpopulations in 106 patients with confirmed SARS-CoV-2 infection who received medical care at our institution. The patients were divided into three groups according to the follow-up date; laboratory analyses of COVID-19 patients were compared with 28 unexposed healthy controls. Regarding B lymphocyte subsets, levels of IgA + CD27+, IgG + CD27+, IgM + CD27- and switched B cells were significantly reduced at the last follow-up compared to unexposed healthy controls (UHC). Of the 106 COVID-19 patients, 56 were clinically classified as featuring PCC. Significant differences between PCC and COVID-19 convalescents compared to UHC were observed in T helper cells and class-switched B cells. However, we did not detect specific or long-lasting immune cellular changes in PCC compared to the non-post-COVID-19 condition.
Collapse
Affiliation(s)
- Luisa Berger
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
| | - Johannes Wolf
- Department of Laboratory Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| | - Sven Kalbitz
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
| | - Nils Kellner
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| | - Christoph Lübbert
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Stephan Borte
- Department of Laboratory Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| |
Collapse
|
8
|
Tang Y, Zou X, Liu P, Dai Y, Wang S, Su X, Yu Y, Tang W, Zhou J, Li C, Mei H, Xiao N, Ou Y, Wang J, Lu G, Lin G, Cheng L. Human umbilical cord-derived mesenchymal stem cell transplantation improves the long COVID. J Med Virol 2024; 96:e29757. [PMID: 38899432 DOI: 10.1002/jmv.29757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
No effective treatments can ameliorate symptoms of long COVID patients. Our study assessed the safety and efficacy of human umbilical cord-derived mesenchymal stem cells (UC-MSCs) in the treatment of long COVID patients. Ten long COVID patients were enrolled and received intravenous infusions of UC-MSCs on Days 0, 7, and 14. Adverse events and clinical symptoms were recorded, and chest-high-resolution CT (HRCT) images and laboratory parameters were analyzed. During UC-MSCs treatment and follow-up, we did not observe serious adverse events, the symptoms of long COVID patients were significantly relieved in a short time, especially sleep difficulty, depression or anxiety, memory issues, and so forth, and the lung lesions were also repaired. The routine laboratory parameters did not exhibit any significant abnormalities following UC-MSCs transplantation (UMSCT). The proportion of regulatory T cells gradually increased, but it was not statistically significant until 12 months. The proportion of naive B cells was elevated, while memory B cells, class-switched B-cells, and nonswitched B-cells decreased at 1 month after infusion. Additionally, we observed a transient elevation in circulating interleukin (IL)-6 after UMSCT, while tumor necrosis factor (TNF)-α, IL-17A, and IL-10 showed no significant changes. The levels of circulating immunoglobulin (Ig) M increased significantly at month 2, while IgA increased significantly at month 6. Furthermore, the SARS-CoV-2 IgG levels remained consistently high in all patients at Month 6, and there was no significant decrease during the subsequent 12-month follow-up. UMSCT was safe and tolerable in long COVID patients. It showed potential in alleviating long COVID symptoms and improving interstitial lung lesions.
Collapse
Affiliation(s)
- Yuling Tang
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiao Zou
- National Engineering Research Center of Human Stem cell, Changsha, China
| | - Ping Liu
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yanni Dai
- National Engineering Research Center of Human Stem cell, Changsha, China
| | - Siqi Wang
- National Engineering Research Center of Human Stem cell, Changsha, China
| | - Xian Su
- National Engineering Research Center of Human Stem cell, Changsha, China
| | - Yan Yu
- National Engineering Research Center of Human Stem cell, Changsha, China
| | - Wenfang Tang
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jia Zhou
- National Engineering Research Center of Human Stem cell, Changsha, China
| | - Chuang Li
- National Engineering Research Center of Human Stem cell, Changsha, China
| | - Hua Mei
- National Engineering Research Center of Human Stem cell, Changsha, China
| | - Na Xiao
- National Engineering Research Center of Human Stem cell, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Yangqi Ou
- National Engineering Research Center of Human Stem cell, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Jian Wang
- National Engineering Research Center of Human Stem cell, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Guangxiu Lu
- National Engineering Research Center of Human Stem cell, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Ge Lin
- National Engineering Research Center of Human Stem cell, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Lamei Cheng
- National Engineering Research Center of Human Stem cell, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
9
|
Xu L, Abudunaibi B, Zeng Z, Zhao Y, Wang Y, Guo X, Zhang Y, Li T, Lu W, Tian W, Guo Z, Su C, Chen T. Relationship of various COVID-19 antibody titer with individual characteristics and prediction of future epidemic trend in Xiamen City, China. J Thorac Dis 2024; 16:2404-2420. [PMID: 38738254 PMCID: PMC11087623 DOI: 10.21037/jtd-23-1516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/29/2024] [Indexed: 05/14/2024]
Abstract
Background Reinfection of coronavirus disease 2019 (COVID-19) has raised concerns about how reliable immunity from infection and vaccination is. With mass testing for the virus halted, understanding the current prevalence of COVID-19 is crucial. This study investigated 1,191 public health workers at the Xiamen Center for Disease Control, focusing on changes in antibody titers and their relationship with individual characteristics. Methods The study began by describing the epidemiological characteristics of the study participants. Multilinear regression (MLR) models were employed to explore the associations between individual attributes and antibody titers. Additionally, group-based trajectory models (GBTMs) were utilized to identify trajectories in antibody titer changes. To predict and simulate future epidemic trends and examine the correlation of antibody decay with epidemics, a high-dimensional transmission dynamics model was constructed. Results Analysis of epidemiological characteristics revealed significant differences in vaccination status between infected and non-infected groups (χ2=376.706, P<0.05). However, the distribution of antibody titers among the infected and vaccinated populations was not significantly different. The MLR model identified age as a common factor affecting titers of immunoglobulin G (IgG), immunoglobulin M (IgM), and neutralizing antibody (NAb), while other factors showed varying impacts. History of pulmonary disease and hospitalization influenced IgG titer, and factors such as gender, smoking, family history of pulmonary diseases, and hospitalization impacted NAb titers. Age was the sole determinant of IgM titers in this study. GBTM analysis indicated a "gradual decline type" trajectory for IgG (95.65%), while IgM and NAb titers remained stable over the study period. The high-dimensional transmission dynamics model predicted and simulated peak epidemic periods in Xiamen City, which correlated with IgG decay. Age-group-specific simulations revealed a higher incidence and infection rate among individuals aged 30-39 years during both the second and third peaks, followed by those aged 40-49, 50-59, 18-29, and 70-79 years. Conclusions Our study shows that antibody titer could be influenced by age, previous pulmonary diseases as well as smoking. Furthermore, the decline in IgG titers is consistent with epidemic trends. These findings emphasize the need for further exploration of these factors and the development of optimized self-protection countermeasures against reinfection.
Collapse
Affiliation(s)
- Liansheng Xu
- Xiamen Center for Disease Control and Prevention, Xiamen, China
| | - Buasiyamu Abudunaibi
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Zhiqi Zeng
- Respiratory Disease AI Laboratory on Epidemic Intelligence and Medical Big Data Instrument Applications, Macao University of Science and Technology, Macao, China
| | - Yunkang Zhao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Yao Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Xiaohao Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Yidun Zhang
- Xiamen Center for Disease Control and Prevention, Xiamen, China
| | - Tao Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Wenkui Lu
- Xiamen Center for Disease Control and Prevention, Xiamen, China
| | - Weiliang Tian
- School of Medicine, Duke University, Durham, NC, USA
| | - Zhinan Guo
- Xiamen Center for Disease Control and Prevention, Xiamen, China
| | - Chenghao Su
- Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, China
| | - Tianmu Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
10
|
Najimi N, Kadi C, Elmtili N, Seghrouchni F, Bakri Y. Unravelling humoral immunity in SARS-CoV-2: Insights from infection and vaccination. Hum Antibodies 2024; 32:85-106. [PMID: 38758995 DOI: 10.3233/hab-230017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Abstract
Following infection and vaccination against SARS-CoV-2, humoral components of the adaptive immune system play a key role in protecting the host. Specifically, B cells generate high-affinity antibodies against various antigens of the virus. In this review, we discuss the mechanisms of immunity initiation through both natural infection and vaccination, shedding light on the activation of B cell subsets in response to SARS-CoV-2 infection and vaccination. The innate immune system serves as the initial line of primary and nonspecific defence against viruses. However, within several days following infection or a vaccine dose, a virus-specific immune response is initiated, primarily by B cells that produce antibodies. These antibodies contribute to the resolution of the disease. Subsequently, these B cells transition into memory B cells, which play a crucial role in providing long-term immunity against the virus. CD4+ T helper cells initiate a cascade, leading to B cell somatic hypermutation, germinal center memory B cells, and the production of neutralizing antibodies. B-cell dysfunction can worsen disease severity and reduce vaccine efficacy. Notably, individuals with B cell immunodeficiency show lower IL-6 production. Furthermore, this review delves into several aspects of immune responses, such as hybrid immunity, which has shown promise in boosting broad-spectrum protection. Cross-reactive immunity is under scrutiny as well, as pre-existing antibodies can offer protection against the disease. We also decipher breakthrough infection mechanisms, especially with the novel variants of the virus. Finally, we discuss some potential therapeutic solutions regarding B cells including convalescent plasma therapy, B-1 cells, B regulatory cell (Breg) modulation, and the use of neutralizing monoclonal antibodies in combating the infection. Ongoing research is crucial to grasp population immunity trends and assess the potential need for booster doses in maintaining effective immune responses against potential viral threats.
Collapse
Affiliation(s)
- Nouhaila Najimi
- Laboratory of Human Pathologies Biology and Center of Genomic of Human Pathologies Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Chaimae Kadi
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
- Laboratory of Biology and Health, Faculty of Sciences of Tétouan, Abdelmalek Essaâdi University, Tétouan, Morocco
| | - Noureddine Elmtili
- Laboratory of Biology and Health, Faculty of Sciences of Tétouan, Abdelmalek Essaâdi University, Tétouan, Morocco
| | - Fouad Seghrouchni
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Youssef Bakri
- Laboratory of Human Pathologies Biology and Center of Genomic of Human Pathologies Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| |
Collapse
|
11
|
D’Orso S, Pirronello M, Verdiani A, Rossini A, Guerrera G, Picozza M, Sambucci M, Misiti A, De Marco L, Salvia A, Caltagirone C, Giardina E, Battistini L, Borsellino G. Primary and Recall Immune Responses to SARS-CoV-2 in Breakthrough Infection. Vaccines (Basel) 2023; 11:1705. [PMID: 38006037 PMCID: PMC10675240 DOI: 10.3390/vaccines11111705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Breakthrough infections in SARS-CoV-2 vaccinated individuals are an ideal circumstance for the simultaneous exploration of both the vaccine-induced memory reaction to the spike (S) protein and the primary response to the membrane (M) and nucleocapsid (N) proteins generated by natural infection. We monitored 15 healthcare workers who had been vaccinated with two doses of Pfizer BioNTech BNT162b2 and were then later infected with the SARS-CoV-2 B.1.617.2. (Delta) variant, analysing the antiviral humoral and cellular immune responses. Natural infection determined an immediate and sharp rise in anti-RBD antibody titres and in the frequency of both S-specific antibody secreting cells (ASCs) and memory B lymphocytes. T cells responded promptly to infection by activating and expanding already at 2-5 days. S-specific memory and emerging M- and N-specific T cells both expressed high levels of activation markers and showed effector capacity with similar kinetics but with different magnitude. The results show that natural infection with SARS-CoV-2 in vaccinated individuals induces fully functional and rapidly expanding T and B lymphocytes in concert with the emergence of novel virus-specific T cells. This swift and punctual response also covers viral variants and captures a paradigmatic case of a healthy adaptive immune reaction to infection with a mutating virus.
Collapse
Affiliation(s)
- Silvia D’Orso
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, 00143 Rome, Italy; (S.D.); (M.P.); (A.V.); (G.G.); (M.P.); (M.S.); (A.M.); (L.D.M.); (L.B.)
| | - Marta Pirronello
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, 00143 Rome, Italy; (S.D.); (M.P.); (A.V.); (G.G.); (M.P.); (M.S.); (A.M.); (L.D.M.); (L.B.)
| | - Alice Verdiani
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, 00143 Rome, Italy; (S.D.); (M.P.); (A.V.); (G.G.); (M.P.); (M.S.); (A.M.); (L.D.M.); (L.B.)
| | - Angelo Rossini
- Medical Services, Santa Lucia Foundation IRCCS, 00179 Rome, Italy; (A.R.); (A.S.)
| | - Gisella Guerrera
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, 00143 Rome, Italy; (S.D.); (M.P.); (A.V.); (G.G.); (M.P.); (M.S.); (A.M.); (L.D.M.); (L.B.)
| | - Mario Picozza
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, 00143 Rome, Italy; (S.D.); (M.P.); (A.V.); (G.G.); (M.P.); (M.S.); (A.M.); (L.D.M.); (L.B.)
| | - Manolo Sambucci
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, 00143 Rome, Italy; (S.D.); (M.P.); (A.V.); (G.G.); (M.P.); (M.S.); (A.M.); (L.D.M.); (L.B.)
| | - Andrea Misiti
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, 00143 Rome, Italy; (S.D.); (M.P.); (A.V.); (G.G.); (M.P.); (M.S.); (A.M.); (L.D.M.); (L.B.)
| | - Lorenzo De Marco
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, 00143 Rome, Italy; (S.D.); (M.P.); (A.V.); (G.G.); (M.P.); (M.S.); (A.M.); (L.D.M.); (L.B.)
| | - Antonino Salvia
- Medical Services, Santa Lucia Foundation IRCCS, 00179 Rome, Italy; (A.R.); (A.S.)
| | - Carlo Caltagirone
- Department of Clinical and Behavioral Neurology, Santa Lucia Foundation IRCCS, 00179 Rome, Italy;
| | - Emiliano Giardina
- Genomic Medicine Laboratory UILDM, Santa Lucia Foundation IRCCS, 00179 Rome, Italy;
- Medical Genetics Laboratory, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Luca Battistini
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, 00143 Rome, Italy; (S.D.); (M.P.); (A.V.); (G.G.); (M.P.); (M.S.); (A.M.); (L.D.M.); (L.B.)
| | - Giovanna Borsellino
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, 00143 Rome, Italy; (S.D.); (M.P.); (A.V.); (G.G.); (M.P.); (M.S.); (A.M.); (L.D.M.); (L.B.)
| |
Collapse
|
12
|
Garcia-Gasalla M, Berman-Riu M, Rodriguez A, Iglesias A, Fraile-Ribot PA, Toledo-Pons N, Pol-Pol E, Ferré-Beltrán A, Artigues-Serra F, Martin-Pena ML, Pons J, Murillas J, Oliver A, Riera M, Ferrer JM. Elevated complement C3 and increased CD8 and type 1 helper lymphocyte T populations in patients with post-COVID-19 condition. Cytokine 2023; 169:156295. [PMID: 37453328 DOI: 10.1016/j.cyto.2023.156295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Biological markers associated to post-COVID-19 condition (PCC) have not been clearly identified. METHODS Eighty-two patients attending our post-COVID-19 outpatient clinic were recruited and classified as fully recovered (40.2%) or presenting with PCC (59.8%). Clinical and radiological data, laboratory markers, cytokines, and lymphocyte populations were analyzed. RESULTS Median number of days after hospitalization was 78.5 [p25-p75: 60-93] days. PCC was significantly more frequent in women, in patients with a previously critical COVID-19, and in those with two or more comorbidities. No differences were found in lymphocyte counts, ferritin, C-reactive protein, D-dimer or sCD25, IL-1β, IL-1Ra, IL-6, CXCL8, IL-17A, IL-18, IL-22, IFN-γ, TNF-α, and IL-10 cytokines levels. PCC patients showed significantly higher levels of complement factor C3 than fully recovered patients: median C3 128 mg/dL [p25-p75:107-135] vs 111 mg/dL [p25-p75: 100-125] (p =.005), respectively. In the flow cytometry assessment of peripheral blood lymphocyte subpopulations, PCC patients showed significantly increased CD8 populations compared to fully recovered patients: median CD8: 529 [p25-p75: 384-683] vs 370/mm3 [p25-p75:280-523], p =.007. When type 1, 2, 17/22, and 17.1 helper and follicular T lymphocyte subpopulations were analyzed, the frequency of Th1 was significantly higher in PCC patients compared to fully recovered patients (30% vs 38.5%, p =.028). CONCLUSION Patients with a post-COVID-19 condition showed significantly increased immunological parameters of inflammation (complement factor C3 and CD8 and Th1 T lymphocyte populations) compared to fully recovered patients. These parameters could be used as biological markers of this condition.
Collapse
Affiliation(s)
- Mercedes Garcia-Gasalla
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain; Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Universitat de les Illes Balears. Palma de Mallorca, Illes Balears, Spain.
| | - Maria Berman-Riu
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Universitat de les Illes Balears. Palma de Mallorca, Illes Balears, Spain
| | - Adrian Rodriguez
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Department of Internal Medicine, Hospital Universitari Son Llàtzer, Palma, Spain
| | - Amanda Iglesias
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Centro de Investigación Biomedica en Red (CIBER) de Enfermedades Respiratorias, Hospital Universitari Son Espases, Palma, Spain
| | - Pablo A Fraile-Ribot
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Department of Microbiology, Hospital Universitari Son Espases, Palma, Spain
| | - Nuria Toledo-Pons
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Department of Pneumology, Hospital Universitari Son Espases, Palma, Spain
| | - Elisabet Pol-Pol
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
| | - Adrian Ferré-Beltrán
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain
| | | | - M Luisa Martin-Pena
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain; Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Jaime Pons
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Centro de Investigación Biomedica en Red (CIBER) de Enfermedades Respiratorias, Hospital Universitari Son Espases, Palma, Spain; Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
| | - Javier Murillas
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain; Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Universitat de les Illes Balears. Palma de Mallorca, Illes Balears, Spain
| | - Antonio Oliver
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Department of Microbiology, Hospital Universitari Son Espases, Palma, Spain; Universitat de les Illes Balears. Palma de Mallorca, Illes Balears, Spain
| | - Melchor Riera
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain; Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Universitat de les Illes Balears. Palma de Mallorca, Illes Balears, Spain
| | - Joana M Ferrer
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain; Department of Immunology, Hospital Universitari Son Espases, Palma, Spain; Universitat de les Illes Balears. Palma de Mallorca, Illes Balears, Spain
| |
Collapse
|
13
|
Castleman MJ, Santos AL, Lesteberg KE, Maloney JP, Janssen WJ, Mould KJ, Beckham JD, Pelanda R, Torres RM. Activation and pro-inflammatory cytokine production by unswitched memory B cells during SARS-CoV-2 infection. Front Immunol 2023; 14:1213344. [PMID: 37638016 PMCID: PMC10449608 DOI: 10.3389/fimmu.2023.1213344] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/03/2023] [Indexed: 08/29/2023] Open
Abstract
Memory B cells are comprised of unswitched (CD27+IgD+) and switched (CD27+IgD-) subsets. The origin and function of unswitched human memory B cells are debated in the literature, whereas switched memory B cells are primed to respond to recurrent infection. Unswitched memory B cells have been described to be reduced in frequency with severe SARS-CoV2 infection and here we characterize their activation status, BCR functionality, and contribution to virally-induced cytokine production. Analyses of whole blood from healthy individuals, people immunized against SARS-CoV2, and those who have had mild and severe SARS-CoV2 infection, confirm a reduction in the frequency of unswitched memory B cells during severe SARS-CoV2 infection and demonstrate this reduction is associated with increased levels of systemic TNFα. We further document how severe viral infection is associated with an increased frequency of 'IgD+' only memory B cells that correlate with increased IgG autoantibody levels. Unswitched and switched memory B cells from severe SARS-CoV2 infection displayed evidence of heightened activation with a concomitant reduction in the expression of the inhibitory receptor CD72. Functionally, both populations of memory B cells from severe SARS-COV2 infection harbored a signaling-competent BCR that displayed enhanced BCR signaling activity in the unswitched population. Finally, we demonstrate that B cells from mild SARS-CoV2 infection are poised to secrete pro-inflammatory cytokines IL-6 and TNFα. Importantly, unswitched memory B cells were a major producer of IL-6 and switched memory B cells were a major producer of TNFα in response to viral TLR ligands. Together these data indicate that B cells contribute to the inflammatory milieu during viral infection.
Collapse
Affiliation(s)
- Moriah J. Castleman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Adriana Luna Santos
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Kelsey E. Lesteberg
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Division of Infectious Disease, University of Colorado School of Medicine, Aurora, CO, United States
| | - James P. Maloney
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - William J. Janssen
- Department of Medicine, National Jewish Health, Denver, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Kara J. Mould
- Department of Medicine, National Jewish Health, Denver, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - J. David Beckham
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Division of Infectious Disease, University of Colorado School of Medicine, Aurora, CO, United States
- Rocky Mountain Regional VA, Medical Center, Aurora, CO, United States
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Raul M. Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
14
|
Castaldo P, d’Alanno G, Biserni GB, Moratti M, Conti F, Fabi M, Lanari M. Exploring Factors Influencing Changes in Incidence and Severity of Multisystem Inflammatory Syndrome in Children. Pathogens 2023; 12:997. [PMID: 37623957 PMCID: PMC10458149 DOI: 10.3390/pathogens12080997] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/16/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023] Open
Abstract
Multisystem inflammatory syndrome (MIS-C) is a rare condition associated with COVID-19 affecting children, characterized by severe and aberrant systemic inflammation leading to nonspecific symptoms, such as gastrointestinal, cardiac, respiratory, hematological, and neurological disorders. In the last year, we have experienced a progressive reduction in the incidence and severity of MIS-C, reflecting the worldwide trend. Thus, starting from the overall trend in the disease in different continents, we reviewed the literature, hypothesizing the potential influencing factors contributing to the reduction in cases and the severity of MIS-C, particularly the vaccination campaign, the spread of different SARS-CoV-2 variants (VOCs), and the changes in human immunological response. The decrease in the severity of MIS-C and its incidence seem to be related to a combination of different factors rather than a single cause. Maturation of an immunological memory to SARS-CoV-2 over time, the implication of mutations of key amino acids of S protein in VOCs, and the overall immune response elicited by vaccination over the loss of neutralization of vaccines to VOCs seem to play an important role in this change.
Collapse
Affiliation(s)
- Pasquale Castaldo
- Specialty School of Pediatrics, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (P.C.); (G.d.); (M.M.)
| | - Gabriele d’Alanno
- Specialty School of Pediatrics, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (P.C.); (G.d.); (M.M.)
| | | | - Mattia Moratti
- Specialty School of Pediatrics, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (P.C.); (G.d.); (M.M.)
| | - Francesca Conti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Marianna Fabi
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.F.); (M.L.)
| | - Marcello Lanari
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.F.); (M.L.)
| |
Collapse
|
15
|
del Pino Molina L, Bravo Gallego LY, Nozal P, Soto-Serrano Y, Martínez-Feito A, Reche-Yebra K, González-Torbay A, Cuesta-Martín de la Cámara R, Gianelli C, Cámara C, González-García J, González-Muñoz M, Rodríguez-Pena R, López Granados E. Detection of specific RBD + IgG + memory B cells by flow cytometry in healthcare workers and patients with inborn errors of immunity after BNT162b2 m RNA COVID-19 vaccination. Front Immunol 2023; 14:1136308. [PMID: 37215146 PMCID: PMC10192857 DOI: 10.3389/fimmu.2023.1136308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/13/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction Inborn errors of immunity (IEI) are a heterogeneous group of diseases caused by intrinsic defects of the immune system. Estimating the immune competence of immunocompromised patients for an infection risk assessment or after SARS-CoV-2 vaccination constituted a challenge. Methods The aim of this study was to determine the humoral responses of patients with IEI through a comprehensive analysis of specific receptor-binding domain-positive (RBD+) IgG+ memory B cells (MBCs) by flow cytometry, together with routine S-specific IgG antibodies and QuantiFERON SARS-CoV-2 (T-cell response), before the vaccine and 3 weeks after a second dose. Results and discussion We first analyzed the percentage of specific RBD+ IgG+ MBCs in healthy healthcare workers. Within the control group, there was an increase in the percentage of specific IgG+ RBD+ MBCs 21 days after the second dose, which was consistent with S-specific IgG antibodies.Thirty-one patients with IEI were included for the pre- and post-vaccination study; IgG+ RBD+ MBCs were not evaluated in 6 patients due to an absence of B cells in peripheral blood. We detected various patterns among the patients with IEI with circulating B cells (25, 81%): an adequate humoral response was observed in 12/25, consider by the detection of positive S-specific IgG antibodies and the presence of specific IgG+ RBD+ MBCs, presenting a positive T-cell response; in 4/25, very low S-specific IgG antibody counts correlated with undetectable events in the IgG+ RBD+ MBC compartment but with positive cellular response. Despite the presence of S-specific IgG antibodies, we were unable to detect a relevant percentage of IgG+ RBD+ MBCs in 5/25; however, all presented positive T-cell response. Lastly, we observed a profound failure of B and T-cell response in 3 (10%) patients with IEI, with no assessment of S-specific IgG antibodies, IgG+ RBD+ MBCs, and negative cellular response. The identification of specific IgG+ RBD+ MBCs by flow cytometry provides information on different humoral immune response outcomes in patients with IEI and aids the assessment of immune competence status after SARS-CoV-2 mRNA vaccine (BNT162b2), together with S-specific IgG antibodies and T-cell responses.
Collapse
Affiliation(s)
- Lucía del Pino Molina
- Center for Biomedical Network Research on Rare Diseases (CIBERER U767), ISCIII, Madrid, Spain
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Luz Yadira Bravo Gallego
- Center for Biomedical Network Research on Rare Diseases (CIBERER U767), ISCIII, Madrid, Spain
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Pilar Nozal
- Clinical Immunology Department, La Paz University Hospital, Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER U754), ISCIII, Madrid, Spain
- Complement Research Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Yolanda Soto-Serrano
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Ana Martínez-Feito
- Clinical Immunology Department, La Paz University Hospital, Madrid, Spain
- Immuno-Rheumatology Research Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Keren Reche-Yebra
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | | | | | - Carla Gianelli
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- Clinical Immunology Department, La Paz University Hospital, Madrid, Spain
| | - Carmen Cámara
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- Clinical Immunology Department, La Paz University Hospital, Madrid, Spain
| | - J. González-García
- HIV Unit, Internal Medicine Department, La Paz University Hospital, AIDS and Infectious Diseases Group, Center for Biomedical Network Research on Infectious Diseases (CIBERINFEC CB21/13/00039), La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | | | - Rebeca Rodríguez-Pena
- Center for Biomedical Network Research on Rare Diseases (CIBERER U767), ISCIII, Madrid, Spain
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- Clinical Immunology Department, La Paz University Hospital, Madrid, Spain
| | - Eduardo López Granados
- Center for Biomedical Network Research on Rare Diseases (CIBERER U767), ISCIII, Madrid, Spain
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- Clinical Immunology Department, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
16
|
Nunez Lopez YO, Iliuk A, Casu A, Parikh A, Smith JS, Corbin K, Lupu D, Pratley RE. Extracellular vesicle proteomics and phosphoproteomics identify pathways for increased risk in patients hospitalized with COVID-19 and type 2 diabetes mellitus. Diabetes Res Clin Pract 2023; 197:110565. [PMID: 36736734 PMCID: PMC9890887 DOI: 10.1016/j.diabres.2023.110565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/22/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
Recent studies suggest that extracellular vesicles (EVs) play a role in the pathogenesis of SARS-CoV-2 infection and the severity of COVID-19. However, their role in the interaction between COVID-19 and type 2 diabetes (T2D) has not been addressed. Here, we characterized the circulating EV proteomic and phosphoproteomic landscape in patients with and without T2D hospitalized with COVID-19 or non-COVID-19 acute respiratory illness (RSP). We detected differentially expressed protein and phosphoprotein signatures that effectively characterized the study groups. The trio of immunomodulatory and coagulation proteins C1QA, C1QB, and C1QC appeared to be a central cluster in both the COVID-19 and T2D functional networks. PKCβ appeared to be retained in cells by being diverted from EV pathways and contribute to the COVID-19 and T2D interaction via a PKC/BTK/TEC axis. EV-shuttled CASP3 and ROCK1 appeared to be coregulated and likely contribute to disease interactions in patients with COVID-19 and T2D. Predicted activation of AMPK, MAPK, and SYK appeared to also play important roles driving disease interaction. These results suggest that activated cellular kinases (i.e., PKC, AMPK, MAPK, and SYK) and multiple EV-shuttled kinases (i.e., PKCβ, BTK, TEC, MAP2K2, and ROCK1) may play key roles in severe COVID-19, particularly in patients with comorbid diabetes.
Collapse
Affiliation(s)
- Yury O Nunez Lopez
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, United States
| | - Anton Iliuk
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, United States; Tymora Analytical Operations, West Lafayette, IN 47906, United States.
| | - Anna Casu
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, United States
| | - Amay Parikh
- Division of Critical Care, AdventHealth Medical Group, AdventHealth Orlando, Orlando, FL 32804, United States
| | - Joshua S Smith
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, United States
| | - Karen Corbin
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, United States
| | - Daniel Lupu
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, United States
| | - Richard E Pratley
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, United States.
| |
Collapse
|
17
|
Velounias RL, Tull TJ. Human B-cell subset identification and changes in inflammatory diseases. Clin Exp Immunol 2022; 210:201-216. [PMID: 36617261 PMCID: PMC9985170 DOI: 10.1093/cei/uxac104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/19/2022] [Accepted: 11/15/2022] [Indexed: 01/09/2023] Open
Abstract
Our understanding of the B-cell subsets found in human blood and their functional significance has advanced greatly in the past decade. This has been aided by the evolution of high dimensional phenotypic tools such as mass cytometry and single-cell RNA sequencing which have revealed heterogeneity in populations that were previously considered homogenous. Despite this, there is still uncertainty and variation between studies as to how B-cell subsets are identified and named. This review will focus on the most commonly encountered subsets of B cells in human blood and will describe gating strategies for their identification by flow and mass cytometry. Important changes to population frequencies and function in common inflammatory and autoimmune diseases will also be described.
Collapse
Affiliation(s)
- Rebekah L Velounias
- Department of Immunobiology, King’s College London, Guy’s Hospital Campus, London, UK
| | - Thomas J Tull
- St John’s Institute of Dermatology, King’s College London, Guy’s Hospital Campus, London, UK
| |
Collapse
|
18
|
Hara A, Chihara N, Akatani R, Nishigori R, Tsuji A, Yoshimura H, Kawamoto M, Otsuka Y, Kageyama Y, Kondo T, Leypoldt F, Wandinger KP, Matsumoto R. Circulating plasmablasts and follicular helper T-cell subsets are associated with antibody-positive autoimmune epilepsy. Front Immunol 2022; 13:1048428. [PMID: 36569937 PMCID: PMC9773883 DOI: 10.3389/fimmu.2022.1048428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/31/2022] [Indexed: 12/13/2022] Open
Abstract
Autoimmune epilepsy (AE) is an inflammatory disease of the central nervous system with symptoms that have seizures that are refractory to antiepileptic drugs. Since the diagnosis of AE tends to rely on a limited number of anti-neuronal antibody tests, a more comprehensive analysis of the immune background could achieve better diagnostic accuracy. This study aimed to compare the characteristics of anti-neuronal antibody-positive autoimmune epilepsy (AE/Ab(+)) and antibody-negative suspected autoimmune epilepsy (AE/Ab(-)) groups. A total of 23 patients who met the diagnostic criteria for autoimmune encephalitis with seizures and 11 healthy controls (HC) were enrolled. All patients were comprehensively analyzed for anti-neuronal antibodies; 13 patients were identified in the AE/Ab(+) group and 10 in the AE/Ab(-) group. Differences in clinical characteristics, including laboratory and imaging findings, were evaluated between the groups. In addition, the immunophenotype of peripheral blood mononuclear cells (PBMCs) and CSF mononuclear cells, particularly B cells and circulating Tfh (cTfh) subsets, and multiplex assays of serum and CSF were analyzed using flow cytometry. Patients with AE/Ab(+) did not show any differences in clinical parameters compared to patients with AE/Ab(-). However, the frequency of plasmablasts within PBMCs and CSF in patients with AE/Ab(+) was higher than that in patients with AE/Ab(-) and HC, and the frequency of cTfh17 cells and inducible T-cell co-stimulator (ICOS) expressing cTfh17 cells within cTfh subsets was higher than that in patients with AE/Ab(-). Furthermore, the frequency of ICOShighcTfh17 cells was positively correlated with that of the unswitched memory B cells. We also found that IL-12, IL-23, IL-6, IL-17A, and IFN-γ levels were elevated in the serum and IL-17A and IL-6 levels were elevated in the CSF of patients with AE/Ab(+). Our findings indicate that patients with AE/Ab(+) showed increased differentiation of B cells and cTfh subsets associated with antibody production. The elevated frequency of plasmablasts and ICOS expressing cTfh17 shift in PBMCs may be indicative of the presence of antibodies in patients with AE.
Collapse
Affiliation(s)
- Atsushi Hara
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Norio Chihara
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Japan,*Correspondence: Norio Chihara, ; Riki Matsumoto,
| | - Ritsu Akatani
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryusei Nishigori
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Asato Tsuji
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hajime Yoshimura
- Department of Neurology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Michi Kawamoto
- Department of Neurology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Yoshihisa Otsuka
- Department of Neurology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Yasufumi Kageyama
- Department of Neurology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Takayuki Kondo
- Department of Neurology, Kansai Medical University Medical Center, Moriguchi, Japan
| | - Frank Leypoldt
- Neuroimmunology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel, Germany,Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Klaus-Peter Wandinger
- Neuroimmunology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Riki Matsumoto
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Japan,*Correspondence: Norio Chihara, ; Riki Matsumoto,
| |
Collapse
|
19
|
Mendez-Cortina Y, Rodriguez-Perea AL, Chvatal-Medina M, Lopera TJ, Alvarez-Mesa N, Rodas-Marín JK, Moncada DC, Rugeles MT, Velilla PA. Dynamics of humoral immune response in SARS-CoV-2 infected individuals with different clinical stages. Front Immunol 2022; 13:1007068. [DOI: 10.3389/fimmu.2022.1007068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
BackgroundThe COVID-19 pandemic remains a global health problem. As in other viral infections, the humoral immune response against SARS-CoV-2 is thought to be crucial for controlling the infection. However, the dynamic of B cells in the clinical spectrum of this disease is still controversial. This study aimed to characterize B cell subsets and neutralizing responses in COVID-19 patients according to disease severity through a one-month follow-up.MethodsA cohort of 71 individuals with SARS-CoV-2 infection confirmed by RT-PCR were recruited and classified into four groups: i) asymptomatic; ii) symptomatic outpatients; iii) hospitalized in ward, and iv) intensive care unit patients (ICU). Samples were taken at days 0 (inclusion to the study), 7 and 30. B cell subsets and neutralizing antibodies were assessed using multiparametric flow cytometry and plaque reduction neutralization, respectively.ResultsOlder age, male gender and body mass index over 25 were common factors among hospitalized and ICU patients, compared to those with milder clinical presentations. In addition, those requiring hospitalization had more comorbidities. A significant increase in the frequencies of CD19+ cells at day 0 was observed in hospitalized and ICU patients compared to asymptomatic and symptomatic groups. Likewise, the frequency of plasmablasts was significantly increased at the first sample in the ICU group compared to the asymptomatic group, but then waned over time. The frequency of naïve B cells decreased at days 7 and 30 compared to day 0 in hospitalized and ICU patients. The neutralizing antibody titers were higher as the severity of COVID-19 increased; in asymptomatic individuals, it was strongly correlated with the percentage of IgM+ switched memory B cells, and a moderate correlation was found with plasmablasts.ConclusionThe humoral immune response is variable among SARS-CoV-2 infected people depending on the severity and time of clinical evolution. In severe COVID-19 patients, a higher plasmablast frequency and neutralizing antibody response were observed, suggesting that, despite having a robust humoral immunity, this response could be late, having a low impact on disease outcome.
Collapse
|
20
|
Network Analysis for Uncovering the Relationship between Host Response and Clinical Factors to Virus Pathogen: Lessons from SARS-CoV-2. Viruses 2022; 14:v14112422. [PMID: 36366522 PMCID: PMC9697085 DOI: 10.3390/v14112422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
Analysing complex datasets while maintaining the interpretability and explainability of outcomes for clinicians and patients is challenging, not only in viral infections. These datasets often include a variety of heterogeneous clinical, demographic, laboratory, and personal data, and it is not a single factor but a combination of multiple factors that contribute to patient characterisation and host response. Therefore, multivariate approaches are needed to analyse these complex patient datasets, which are impossible to analyse with univariate comparisons (e.g., one immune cell subset versus one clinical factor). Using a SARS-CoV-2 infection as an example, we employed a patient similarity network (PSN) approach to assess the relationship between host immune factors and the clinical course of infection and performed visualisation and data interpretation. A PSN analysis of ~85 immunological (cellular and humoral) and ~70 clinical factors in 250 recruited patients with coronavirus disease (COVID-19) who were sampled four to eight weeks after a PCR-confirmed SARS-CoV-2 infection identified a minimal immune signature, as well as clinical and laboratory factors strongly associated with disease severity. Our study demonstrates the benefits of implementing multivariate network approaches to identify relevant factors and visualise their relationships in a SARS-CoV-2 infection, but the model is generally applicable to any complex dataset.
Collapse
|
21
|
Çölkesen F, Kepenek Kurt E, Vatansev H, Korkmaz C, Çölkesen F, Yücel F, Yıldız E, Evcen R, Aykan FS, Kılınç M, Aytekin G, Feyzioğlu B, Doğan M, Arslan Ş, Teke T, Keleş S, Reisli İ. Memory B cells and serum immunoglobulins are associated with disease severity and mortality in patients with COVID-19. Postgrad Med J 2022; 98:765-771. [PMID: 37062997 PMCID: PMC8783971 DOI: 10.1136/postgradmedj-2021-140540] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 01/06/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE OF THE STUDY The aim of this study was to investigate the relationship of B cell-mediated immunity with disease severity and mortality in patients with COVID-19. STUDY DESIGN In this retrospective cohort and single-centre study, 208 patients with laboratory-confirmed COVID-19 were recruited. A COVID-19 severity score, ranging from 0 to 10, was used to evaluate associations between various factors. Serum immunoglobulin levels and the number of cells in B lymphocyte subsets were measured and their association with disease severity and mortality in patients with COVID-19 examined. RESULTS The median age of the patients was 50 (35-63) years and 88 (42%) were female. The number of deceased patients was 17. The median COVID-19 severity score was 8 (6-8) in deceased patients and 1 (0-2) in survivors. Deceased patients had significantly lower levels of total B lymphocytes, naive B cells, switched memory B cells, and serum IgA, IgG, IgG1 and IgG2 than recovered patients (all p<0.05). In addition, a significant negative correlation was found between the number of these parameters and COVID-19 severity scores. Decrease in the number of total B cells and switched memory B cells as well as lower serum IgA, IgG and IgG1 levels were independent risk factors for mortality in patients with COVID-19. CONCLUSION In the present study, the prognosis of patients with COVID-19 was shown to be associated with the B cell subset and serum immunoglobulin levels.
Collapse
Affiliation(s)
- Fatih Çölkesen
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Esma Kepenek Kurt
- Department of Infectious Diseases and Clinical Microbiology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Hülya Vatansev
- Department of Chest Diseases, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Celalettin Korkmaz
- Department of Chest Diseases, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Fatma Çölkesen
- Department of Infectious Diseases and Clinical Microbiology, Konya Training and Research Hospital, Konya, Turkey
| | - Fatih Yücel
- Department of General Intensive Care Unit, Konya Training and Research Hospital, Konya, Turkey
| | - Eray Yıldız
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Recep Evcen
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Filiz Sadi Aykan
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Mehmet Kılınç
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Gökhan Aytekin
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Konya Training and Research Hospital, Konya, Turkey
| | - Bahadır Feyzioğlu
- Division of Medical Virology, Department of Medical Microbiology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Metin Doğan
- Department of Medical Microbiology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Şevket Arslan
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Turgut Teke
- Department of Chest Diseases, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Sevgi Keleş
- Division of Pediatric Allergy and Immunology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - İsmail Reisli
- Division of Pediatric Allergy and Immunology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| |
Collapse
|
22
|
Maamari KA, Busaidi IA, Kindi MA, Zadjali F, BaAlawi F, Anesta W, Amri KA, Albalushi W, Balushi HA, Amri AA, Aljufaili M, Al-Busaidi M, Muharrmi ZA, Balkhair A, Riyami NA, Ghanim Z, Alshekaili J. Short and long-term immune changes in different severity groups of COVID-19 disease. Int J Infect Dis 2022; 122:776-784. [PMID: 35840099 PMCID: PMC9284586 DOI: 10.1016/j.ijid.2022.07.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND There are limited data on short- versus long-term changes in adaptive immune response across different COVID-19 disease severity groups. METHODS A multicenter prospective study of 140 adult patients with COVID-19 (a total of 325 samples) were analyzed for inflammatory markers and lymphocyte subsets at presentation, week 2, and week 24. RESULTS Inflammatory markers at presentation were higher in the critical/severe than in moderate and mild groups. A predominance of memory B cell response in the mild and moderate group was noted by week 2. In contrast, the immune system in the severe/critical group was dysfunctional, with expansion of exhausted CD8+ T cells and atypical memory B cells. By 24 weeks, there was a possible trend of normalization. CONCLUSION There was substantial difference in the degree of inflammation and distribution of different B and T cell subsets in the different disease severity groups. Despite the initial dysfunctional immune response in the severe/critical group, a comparable memory B and CD8+ T cell responses to the mild group was achieved at 24 weeks.
Collapse
Affiliation(s)
- Khuloud Al Maamari
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Ibrahim Al Busaidi
- Department of Medicine, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Mahmood Al Kindi
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Fahad Zadjali
- Department of Clinical Biochemistry, Sultan Qaboos University, Sultanate of Oman
| | - Fatma BaAlawi
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Wijesinghe Anesta
- Department of internal medicine, Armed Forces Hospital, Sultanate of Oman
| | - Kawthar Al Amri
- Department of internal medicine, Armed Forces Hospital, Sultanate of Oman
| | - Wafa Albalushi
- Department of Nursing, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Hamed Al Balushi
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Ayman Al Amri
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Mahmood Aljufaili
- Department of Emergency, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Mujahid Al-Busaidi
- Department of Medicine, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Zakariya Al Muharrmi
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Abdullah Balkhair
- Department of Medicine, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Nafila Al Riyami
- Department of Clinical Biochemistry, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Zahraa Ghanim
- Department of Anaesthesia, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Jalila Alshekaili
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman,Corresponding author
| |
Collapse
|
23
|
Emmenegger M, Fiedler S, Brugger SD, Devenish SR, Morgunov AS, Ilsley A, Ricci F, Malik AY, Scheier T, Batkitar L, Madrigal L, Rossi M, Meisl G, Lynn AK, Saleh L, von Eckardstein A, Knowles TP, Aguzzi A. Both COVID-19 infection and vaccination induce high-affinity cross-clade responses to SARS-CoV-2 variants. iScience 2022; 25:104766. [PMID: 35875683 PMCID: PMC9288251 DOI: 10.1016/j.isci.2022.104766] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/06/2022] [Accepted: 07/08/2022] [Indexed: 11/20/2022] Open
Abstract
The B.1.1.529 (omicron) variant has rapidly supplanted most other SARS-CoV-2 variants. Using microfluidics-based antibody affinity profiling (MAAP), we have characterized affinity and IgG concentration in the plasma of 39 individuals with multiple trajectories of SARS-CoV-2 infection and/or vaccination. Antibody affinity was similar against the wild-type, delta, and omicron variants (K A ranges: 122 ± 155, 159 ± 148, 211 ± 307 μM-1, respectively), indicating a surprisingly broad and mature cross-clade immune response. Postinfectious and vaccinated subjects showed different IgG profiles, with IgG3 (p-value = 0.002) against spike being more prominent in the former group. Lastly, we found that the ELISA titers correlated linearly with measured concentrations (R = 0.72) but not with affinity (R = 0.29). These findings suggest that the wild-type and delta spike induce a polyclonal immune response capable of binding the omicron spike with similar affinity. Changes in titers were primarily driven by antibody concentration, suggesting that B-cell expansion, rather than affinity maturation, dominated the response after infection or vaccination.
Collapse
Affiliation(s)
- Marc Emmenegger
- Institute of Neuropathology, University of Zurich, 8091 Zurich, Switzerland
| | - Sebastian Fiedler
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Silvio D. Brugger
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sean R.A. Devenish
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Alexey S. Morgunov
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Alison Ilsley
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Francesco Ricci
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Anisa Y. Malik
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Thomas Scheier
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Leyla Batkitar
- Institute of Neuropathology, University of Zurich, 8091 Zurich, Switzerland
| | - Lidia Madrigal
- Institute of Neuropathology, University of Zurich, 8091 Zurich, Switzerland
| | - Marco Rossi
- Department of Laboratory Medicine, University Hospital Zürich, 8091 Zurich, Switzerland
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Andrew K. Lynn
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Lanja Saleh
- Department of Laboratory Medicine, University Hospital Zürich, 8091 Zurich, Switzerland
| | | | - Tuomas P.J. Knowles
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, JJ Thomson Ave, Cambridge CB3 0HE, UK
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
24
|
Hale M, Netland J, Chen Y, Thouvenel CD, Smith KN, Rich LM, Vanderwall ER, Miranda MC, Eggenberger J, Hao L, Watson MJ, Mundorff CC, Rodda LB, King NP, Guttman M, Gale M, Abraham J, Debley JS, Pepper M, Rawlings DJ. IgM antibodies derived from memory B cells are potent cross-variant neutralizers of SARS-CoV-2. J Exp Med 2022; 219:213384. [PMID: 35938988 PMCID: PMC9365875 DOI: 10.1084/jem.20220849] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/22/2022] [Accepted: 07/12/2022] [Indexed: 01/14/2023] Open
Abstract
Humoral immunity to SARS-CoV-2 can be supplemented with polyclonal sera from convalescent donors or an engineered monoclonal antibody (mAb) product. While pentameric IgM antibodies are responsible for much of convalescent sera's neutralizing capacity, all available mAbs are based on the monomeric IgG antibody subtype. We now show that IgM mAbs derived from immune memory B cell receptors are potent neutralizers of SARS-CoV-2. IgM mAbs outperformed clonally identical IgG antibodies across a range of affinities and SARS-CoV-2 receptor-binding domain epitopes. Strikingly, efficacy against SARS-CoV-2 viral variants was retained for IgM but not for clonally identical IgG. To investigate the biological role for IgM memory in SARS-CoV-2, we also generated IgM mAbs from antigen-experienced IgM+ memory B cells in convalescent donors, identifying a potent neutralizing antibody. Our results highlight the therapeutic potential of IgM mAbs and inform our understanding of the role for IgM memory against a rapidly mutating pathogen.
Collapse
Affiliation(s)
- Malika Hale
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA
| | - Jason Netland
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Yu Chen
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA
| | | | | | - Lucille M. Rich
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA
| | | | - Marcos C. Miranda
- Institute for Protein Design, University of Washington, Seattle, WA,Department of Biochemistry, University of Washington School of Medicine, Seattle, WA
| | - Julie Eggenberger
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Linhui Hao
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Michael J. Watson
- Department of Medicinal Chemistry, University of Washington, Seattle, WA
| | | | - Lauren B. Rodda
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Neil P. King
- Institute for Protein Design, University of Washington, Seattle, WA,Department of Biochemistry, University of Washington School of Medicine, Seattle, WA
| | - Miklos Guttman
- Department of Medicinal Chemistry, University of Washington, Seattle, WA
| | - Michael Gale
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Jonathan Abraham
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA
| | - Jason S. Debley
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA
| | - Marion Pepper
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - David J. Rawlings
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA,Department of Immunology, University of Washington School of Medicine, Seattle, WA,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA,Correspondence to David J. Rawlings:
| |
Collapse
|
25
|
Peng L, Fang Z, Renauer PA, McNamara A, Park JJ, Lin Q, Zhou X, Dong MB, Zhu B, Zhao H, Wilen CB, Chen S. Multiplexed LNP-mRNA vaccination against pathogenic coronavirus species. Cell Rep 2022; 40:111160. [PMID: 35921835 PMCID: PMC9294034 DOI: 10.1016/j.celrep.2022.111160] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/07/2022] [Accepted: 07/13/2022] [Indexed: 12/02/2022] Open
Abstract
Although COVID-19 vaccines have been developed, multiple pathogenic coronavirus species exist, urging on development of multispecies coronavirus vaccines. Here we develop prototype lipid nanoparticle (LNP)-mRNA vaccine candidates against SARS-CoV-2 Delta, SARS-CoV, and MERS-CoV, and we test how multiplexing LNP-mRNAs can induce effective immune responses in animal models. Triplex and duplex LNP-mRNA vaccinations induce antigen-specific antibody responses against SARS-CoV-2, SARS-CoV, and MERS-CoV. Single-cell RNA sequencing profiles the global systemic immune repertoires and respective transcriptome signatures of vaccinated animals, revealing a systemic increase in activated B cells and differential gene expression across major adaptive immune cells. Sequential vaccination shows potent antibody responses against all three species, significantly stronger than simultaneous vaccination in mixture. These data demonstrate the feasibility, antibody responses, and single-cell immune profiles of multispecies coronavirus vaccination. The direct comparison between simultaneous and sequential vaccination offers insights into optimization of vaccination schedules to provide broad and potent antibody immunity against three major pathogenic coronavirus species.
Collapse
Affiliation(s)
- Lei Peng
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA
| | - Zhenhao Fang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA
| | - Paul A Renauer
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA; Molecular Cell Biology, Genetics and Development Program, Yale University, New Haven, CT 06516, USA
| | - Andrew McNamara
- Department of Immunobiology, Yale University, New Haven, CT 06510, USA; Department of Laboratory Medicine, Yale University, New Haven, CT 06510, USA
| | - Jonathan J Park
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA; M.D.-Ph.D. Program, Yale University, West Haven, CT 06516, USA
| | - Qianqian Lin
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA
| | - Xiaoyu Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA
| | - Matthew B Dong
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA; Department of Immunobiology, Yale University, New Haven, CT 06510, USA; M.D.-Ph.D. Program, Yale University, West Haven, CT 06516, USA; Immunobiology Program, Yale University, New Haven, CT 06510, USA
| | - Biqing Zhu
- Computational Biology and Bioinformatics Program, Yale University, New Haven, CT 06510, USA
| | - Hongyu Zhao
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Computational Biology and Bioinformatics Program, Yale University, New Haven, CT 06510, USA; Department of Biostatistics, Yale University School of Public Health, New Haven, CT 06510, USA; Yale Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Craig B Wilen
- Department of Immunobiology, Yale University, New Haven, CT 06510, USA; Department of Laboratory Medicine, Yale University, New Haven, CT 06510, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA; Molecular Cell Biology, Genetics and Development Program, Yale University, New Haven, CT 06516, USA; M.D.-Ph.D. Program, Yale University, West Haven, CT 06516, USA; Immunobiology Program, Yale University, New Haven, CT 06510, USA; Computational Biology and Bioinformatics Program, Yale University, New Haven, CT 06510, USA; Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
26
|
Tertel T, Tomić S, Đokić J, Radojević D, Stevanović D, Ilić N, Giebel B, Kosanović M. Serum-derived extracellular vesicles: Novel biomarkers reflecting the disease severity of COVID-19 patients. J Extracell Vesicles 2022; 11:e12257. [PMID: 35979935 PMCID: PMC9451525 DOI: 10.1002/jev2.12257] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/10/2022] [Accepted: 07/26/2022] [Indexed: 11/09/2022] Open
Abstract
COVID-19 is characterized by a wide spectrum of disease severity, whose indicators and underlying mechanisms need to be identified. The role of extracellular vesicles (EVs) in COVID-19 and their biomarker potential, however, remains largely unknown. Aiming to identify specific EV signatures of patients with mild compared to severe COVID-19, we characterized the EV composition of 20 mild and 26 severe COVID-19 patients along with 16 sex and age-matched healthy donors with a panel of eight different antibodies by imaging flow cytometry (IFCM). We correlated the obtained data with 37 clinical, prerecorded biochemical and immunological parameters. Severe patients' sera contained increased amounts of CD13+ and CD82+ EVs, which positively correlated with IL-6-producing and circulating myeloid-derived suppressor cells (MDSCs) and with the serum concentration of proinflammatory cytokines, respectively. Sera of mild COVID-19 patients contained more HLA-ABC+ EVs than sera of the healthy donors and more CD24+ EVs than severe COVID-19 patients. Their increased abundance negatively correlated with disease severity and accumulation of MDSCs, being considered as key drivers of immunopathogenesis in COVID-19. Altogether, our results support the potential of serum EVs as powerful biomarkers for COVID-19 severity and pave the way for future investigations aiming to unravel the role of EVs in COVID-19 progression.
Collapse
Affiliation(s)
- Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sergej Tomić
- Institute for the Application of Nuclear Energy, INEP, University of Belgrade, Zemun-Belgrade, Serbia
| | - Jelena Đokić
- Institute of Molecular Genetics and Genetic Engineering (IMGGI), University of Belgrade, Belgrade, Serbia
| | - Dušan Radojević
- Institute of Molecular Genetics and Genetic Engineering (IMGGI), University of Belgrade, Belgrade, Serbia
| | - Dejan Stevanović
- Clinical Hospital Center Zemun, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nataša Ilić
- Institute for the Application of Nuclear Energy, INEP, University of Belgrade, Zemun-Belgrade, Serbia
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Maja Kosanović
- Institute for the Application of Nuclear Energy, INEP, University of Belgrade, Zemun-Belgrade, Serbia
| |
Collapse
|
27
|
Jonigk D, Werlein C, Acker T, Aepfelbacher M, Amann KU, Baretton G, Barth P, Bohle RM, Büttner A, Büttner R, Dettmeyer R, Eichhorn P, Elezkurtaj S, Esposito I, Evert K, Evert M, Fend F, Gaßler N, Gattenlöhner S, Glatzel M, Göbel H, Gradhand E, Hansen T, Hartmann A, Heinemann A, Heppner FL, Hilsenbeck J, Horst D, Kamp JC, Mall G, Märkl B, Ondruschka B, Pablik J, Pfefferle S, Quaas A, Radbruch H, Röcken C, Rosenwald A, Roth W, Rudelius M, Schirmacher P, Slotta-Huspenina J, Smith K, Sommer L, Stock K, Ströbel P, Strobl S, Titze U, Weirich G, Weis J, Werner M, Wickenhauser C, Wiech T, Wild P, Welte T, von Stillfried S, Boor P. Organ manifestations of COVID-19: what have we learned so far (not only) from autopsies? Virchows Arch 2022; 481:139-159. [PMID: 35364700 PMCID: PMC8975445 DOI: 10.1007/s00428-022-03319-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 01/08/2023]
Abstract
The use of autopsies in medicine has been declining. The COVID-19 pandemic has documented and rejuvenated the importance of autopsies as a tool of modern medicine. In this review, we discuss the various autopsy techniques, the applicability of modern analytical methods to understand the pathophysiology of COVID-19, the major pathological organ findings, limitations or current studies, and open questions. This article summarizes published literature and the consented experience of the nationwide network of clinical, neuro-, and forensic pathologists from 27 German autopsy centers with more than 1200 COVID-19 autopsies. The autopsy tissues revealed that SARS-CoV-2 can be found in virtually all human organs and tissues, and the majority of cells. Autopsies have revealed the organ and tissue tropism of SARS-CoV-2, and the morphological features of COVID-19. This is characterized by diffuse alveolar damage, combined with angiocentric disease, which in turn is characterized by endothelial dysfunction, vascular inflammation, (micro-) thrombosis, vasoconstriction, and intussusceptive angiogenesis. These findings explained the increased pulmonary resistance in COVID-19 and supported the recommendations for antithrombotic treatment in COVID-19. In contrast, in extra-respiratory organs, pathological changes are often nonspecific and unclear to which extent these changes are due to direct infection vs. indirect/secondary mechanisms of organ injury, or a combination thereof. Ongoing research using autopsies aims at answering questions on disease mechanisms, e.g., focusing on variants of concern, and future challenges, such as post-COVID conditions. Autopsies are an invaluable tool in medicine and national and international interdisciplinary collaborative autopsy-based research initiatives are essential.
Collapse
Affiliation(s)
- Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany.
| | | | - Till Acker
- Institute of Neuropathology, University Hospital Giessen and Marburg, Giessen, Germany
| | - Martin Aepfelbacher
- Institute of Medical Microbiology, Virology, and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin U Amann
- Department of Nephropathology, University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Gustavo Baretton
- Department of Pathology, University Hospital Dresden, Dresden, Germany
| | - Peter Barth
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Rainer M Bohle
- Department of Pathology, University Hospital Saarland Homburg, Homburg, Germany
| | - Andreas Büttner
- Institute of Forensic Medicine, University Medical Center Rostock, Rostock, Germany
| | - Reinhard Büttner
- Department of Pathology, University Hospital Cologne, Cologne, Germany
| | - Reinhard Dettmeyer
- Department of Legal Medicine, University Hospital Giessen and Marburg, Giessen, Germany
| | - Philip Eichhorn
- Department of Pathology, University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Sefer Elezkurtaj
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Irene Esposito
- Department of Pathology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Katja Evert
- Department of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Evert
- Department of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - Falko Fend
- Department of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Nikolaus Gaßler
- Department of Pathology, University Hospital Jena, Jena, Germany
| | - Stefan Gattenlöhner
- Department of Pathology, University Hospital Giessen and Marburg, Giessen, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Heike Göbel
- Department of Pathology, University Hospital Cologne, Cologne, Germany
| | - Elise Gradhand
- Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt, Germany
| | - Torsten Hansen
- Department of Pathology, University Hospital OWL of the Bielefeld University, Campus Lippe, Detmold, Germany
| | - Arndt Hartmann
- Department of Pathology, University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Axel Heinemann
- Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank L Heppner
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
- Cluster of Excellence, NeuroCure, Berlin, Germany
| | - Julia Hilsenbeck
- Department of Pathology, University Hospital Dresden, Dresden, Germany
| | - David Horst
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jan C Kamp
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - Gita Mall
- Department of Legal Medicine, University Hospital Jena, Jena, Germany
| | - Bruno Märkl
- General Pathology and Molecular Diagnostics, University Hospital Augsburg, Augsburg, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jessica Pablik
- Department of Pathology, University Hospital Dresden, Dresden, Germany
| | - Susanne Pfefferle
- Institute of Medical Microbiology, Virology, and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander Quaas
- Department of Pathology, University Hospital Cologne, Cologne, Germany
| | - Helena Radbruch
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christoph Röcken
- Department of Pathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Wilfried Roth
- Department of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Martina Rudelius
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Peter Schirmacher
- Department of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Julia Slotta-Huspenina
- Department of Pathology, TUM School of Medicine of Technical University of Munich, Munich, Germany
| | - Kevin Smith
- Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt, Germany
| | - Linna Sommer
- Department of Pathology, University Hospital Dresden, Dresden, Germany
| | - Konrad Stock
- Department of Nephrology, TUM School of Medicine of Technical University of Munich, Munich, Germany
| | - Philipp Ströbel
- Department of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Stephanie Strobl
- Department of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Ulf Titze
- Department of Pathology, University Hospital OWL of the Bielefeld University, Campus Lippe, Detmold, Germany
| | - Gregor Weirich
- Department of Pathology, TUM School of Medicine of Technical University of Munich, Munich, Germany
| | - Joachim Weis
- Department of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Martin Werner
- Institute for Surgical Pathology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Claudia Wickenhauser
- Department of Pathology, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Thorsten Wiech
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Wild
- Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | | | - Peter Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany.
- Department of Nephrology and Immunology, University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|
28
|
Rossi CM, Lenti MV, Merli S, Di Sabatino A. Role of IgM Memory B Cells and Spleen Function in COVID-19. Front Immunol 2022; 13:889876. [PMID: 35844543 PMCID: PMC9280616 DOI: 10.3389/fimmu.2022.889876] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
IgM memory B cells, are a peculiar subset of memory B cells, which probably originates in the spleen and outside germinal centers and provide a rapid line of defence against mucosal infections. Their role in counteracting COVID-19 is still elusive but, recent evidence, mainly boosted by studies on spleen function/involvement in COVID-19, seems to support the notion that this subset of memory B cells could exert a protective role against this virus, along with other coronaviruses, particularly in the acute setting of the infection, as outlined by worst clinical outcomes observed in unvaccinated patients with impaired IgM B memory response and spleen function. Herein we critically summarise the current landscape of studies on IgM memory B cells, focusing on the clinical impact of their depletion, by comparing the COVID-19-related splenic dysfunction with other hypo- and asplenic conditions and by adding recent data on follow-up studies and postulate a mechanistic explanation for their reduced numbers. The early detection of an impaired IgM memory B cell response in patients with COVID-19 may contribute to their improved care through different strategies, such as through tailored vaccine strategies, prompt hospital admission and/or administration of anti-infective treatments, thus resulting in an better prognosis, although at present management algorithms are still unavailable. Moreover, further studies with longer follow-up are needed to assess the evolution of COVID-19-associated/exacerbated immune deficit.
Collapse
|
29
|
Newell KL, Waldran MJ, Thomas SJ, Endy TP, Waickman AT. Simultaneous analysis of antigen-specific B and T cells after SARS-CoV-2 infection and vaccination. Cytometry A 2022; 101:474-482. [PMID: 35468250 PMCID: PMC9087145 DOI: 10.1002/cyto.a.24563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/10/2022] [Accepted: 04/22/2022] [Indexed: 11/30/2022]
Abstract
Conventional methods for quantifying and phenotyping antigen-specific lymphocytes can rapidly deplete irreplaceable specimens. This is due to the fact that antigen-specific T and B cells have historically been analyzed in independent assays each requiring millions of cells. A technique that facilitates the simultaneous detection of antigen-specific T and B cells would allow for more thorough immune profiling with significantly reduced sample requirements. To this end, we developed the B and T cell tandem lymphocyte evaluation (BATTLE) assay, which allows for the simultaneous identification of SARS-CoV-2 Spike reactive T and B cells using an activation induced marker (AIM) T cell assay and dual-color B cell antigen probes. Using this assay, we demonstrate that antigen-specific B and T cell subsets can be identified simultaneously using conventional flow cytometry platforms and provide insight into the differential effects of mRNA vaccination on B and T cell populations following natural SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Krista L. Newell
- Department of Microbiology and ImmunologySUNY Upstate Medical UniversitySyracuseNew YorkUSA
| | - Mitchell J. Waldran
- Department of Microbiology and ImmunologySUNY Upstate Medical UniversitySyracuseNew YorkUSA
| | - Stephen J. Thomas
- Department of Microbiology and ImmunologySUNY Upstate Medical UniversitySyracuseNew YorkUSA
- Institute for Global Health and Translational ScienceSUNY Upstate Medical UniversitySyracuseNew YorkUSA
| | - Timothy P. Endy
- Department of Microbiology and ImmunologySUNY Upstate Medical UniversitySyracuseNew YorkUSA
| | - Adam T. Waickman
- Department of Microbiology and ImmunologySUNY Upstate Medical UniversitySyracuseNew YorkUSA
- Institute for Global Health and Translational ScienceSUNY Upstate Medical UniversitySyracuseNew YorkUSA
| |
Collapse
|
30
|
Carril-Ajuria L, Desnoyer A, Meylan M, Dalban C, Naigeon M, Cassard L, Vano Y, Rioux-Leclercq N, Chouaib S, Beuselinck B, Chabaud S, Barros-Monteiro J, Bougoüin A, Lacroix G, Colina-Moreno I, Tantot F, Boselli L, De Oliveira C, Fridman WH, Escudier B, Sautes-Fridman C, Albiges L, Chaput-Gras N. Baseline circulating unswitched memory B cells and B-cell related soluble factors are associated with overall survival in patients with clear cell renal cell carcinoma treated with nivolumab within the NIVOREN GETUG-AFU 26 study. J Immunother Cancer 2022; 10:jitc-2022-004885. [PMID: 35640928 PMCID: PMC9157347 DOI: 10.1136/jitc-2022-004885] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The phase II NIVOREN GETUG-AFU 26 study reported safety and efficacy of nivolumab in patients with metastatic clear cell renal cell carcinoma (m-ccRCC) in a 'real-world setting'. We conducted a translational-research program to determine whether specific circulating immune-cell populations and/or soluble factors at baseline were predictive of clinical outcomes in patients with m-ccRCC treated with nivolumab within the NIVOREN study. METHODS Absolute numbers of 106 circulating immune-cell populations were prospectively analyzed in patients treated at a single institution within the NIVOREN trial with available fresh-whole-blood, using dry formulation panels for multicolor flow cytometry. In addition, a panel of 14 predefined soluble factors was quantified for each baseline plasma sample using the Meso-Scale-Discovery immunoassay. The remaining patients with available plasma sample were used as a validation cohort for the soluble factor quantification analysis. Tumor immune microenvironment characterization of all patients included in the translational program of the study was available. The association of blood and tissue-based biomarkers, with overall survival (OS), progression-free survival (PFS) and response was analyzed. RESULTS Among the 44 patients, baseline unswitched memory B cells (NSwM B cells) were enriched in responders (p=0.006) and associated with improved OS (HR=0.08, p=0.002) and PFS (HR=0.54, p=0.048). Responders were enriched in circulating T follicular helper (Tfh) (p=0.027) and tertiary lymphoid structures (TLS) (p=0.043). Circulating NSwM B cells positively correlated with Tfh (r=0.70, p<0.001). Circulating NSwM B cells correlated positively with TLS and CD20 +B cells at the tumor center (r=0.59, p=0.044, and r=0.52, p=0.033) and inversely correlated with BCA-1/CXCL13 and BAFF (r=-0.55 and r=-0.42, p<0.001). Tfh cells also inversely correlated with BCA-1/CXCL13 (r=-0.61, p<0.001). IL-6, BCA-1/CXCL13 and BAFF significantly associated with worse OS in the discovery (n=40) and validation cohorts (n=313). CONCLUSION We report the first fresh blood immune-monitoring of patients with m-ccRCC treated with nivolumab. Baseline blood concentration of NSwM B cells was associated to response, PFS and OS in patients with m-ccRCC treated with nivolumab. BCA-1/CXCL13 and BAFF, inversely correlated to NSwM B cells, were both associated with worse OS in discovery and validation cohorts. Our data confirms a role for B cell subsets in the response to immune checkpoint blockade therapy in patients with m-ccRCC. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Lucia Carril-Ajuria
- Department of Cancer Medicine, Institut Gustave-Roussy, Villejuif, France.,Laboratory for Immunomonitoring in Oncology, Institut Gustave-Roussy, Villejuif, France
| | - Aude Desnoyer
- Laboratory for Immunomonitoring in Oncology, Institut Gustave-Roussy, Villejuif, France.,Faculté de Pharmacie, Université Paris-Saclay, Chatenay-Malabray, France
| | - Maxime Meylan
- Centre de Recherche des Cordeliers, Inserm UMR S1138, Paris, France
| | - Cécile Dalban
- Department of Biostatistics, Centre Leon Bernard, Lyon, France
| | - Marie Naigeon
- Faculté de Pharmacie, Université Paris-Saclay, Chatenay-Malabray, France.,Laboratoire d'immunomonitoring En Oncologie, Institut Gustave-Roussy, Villejuif, France.,Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicetre, France
| | - Lydie Cassard
- Laboratory for Immunomonitoring in Oncology, Institut Gustave-Roussy, Villejuif, France
| | - Yann Vano
- Centre de Recherche des Cordeliers, Inserm UMR S1138, Paris, France.,Service d'Oncologie Medicale, Hopital Europeen Georges Pompidou, Paris, France
| | - Nathalie Rioux-Leclercq
- Service Anatomie Etcytologie Pathologiques, CHU, Université de Rennes, Universite de Rennes 1, Rennes, France
| | - Salem Chouaib
- Department of Immunology, Gustave Roussy Institute, Villejuif, France
| | | | - Sylvie Chabaud
- Department of Biostatistics, Centre Leon Bernard, Lyon, France
| | | | - Antoine Bougoüin
- Centre de Recherche des Cordeliers, Inserm UMR S1138, Paris, France
| | | | | | | | - Lisa Boselli
- Laboratory for Immunomonitoring in Oncology, Institut Gustave-Roussy, Villejuif, France
| | - Caroline De Oliveira
- Laboratory for Immunomonitoring in Oncology, Institut Gustave-Roussy, Villejuif, France
| | | | - Bernard Escudier
- Department of Cancer Medicine, Institut Gustave-Roussy, Villejuif, France
| | | | - Laurence Albiges
- Department of Cancer Medicine, Institut Gustave-Roussy, Villejuif, France.,Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicetre, France
| | - Nathalie Chaput-Gras
- Laboratory for Immunomonitoring in Oncology, Institut Gustave-Roussy, Villejuif, France .,Faculté de Pharmacie, Université Paris-Saclay, Chatenay-Malabray, France
| |
Collapse
|
31
|
Kudryavtsev I, Rubinstein A, Golovkin A, Kalinina O, Vasilyev K, Rudenko L, Isakova-Sivak I. Dysregulated Immune Responses in SARS-CoV-2-Infected Patients: A Comprehensive Overview. Viruses 2022; 14:1082. [PMID: 35632823 PMCID: PMC9147674 DOI: 10.3390/v14051082] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first detected in humans more than two years ago and caused an unprecedented socio-economic burden on all countries around the world. Since then, numerous studies have attempted to identify various mechanisms involved in the alterations of innate and adaptive immunity in COVID-19 patients, with the ultimate goal of finding ways to correct pathological changes and improve disease outcomes. State-of-the-art research methods made it possible to establish precise molecular mechanisms which the new virus uses to trigger multisystem inflammatory syndrome and evade host antiviral immune responses. In this review, we present a comprehensive analysis of published data that provide insight into pathological changes in T and B cell subsets and their phenotypes, accompanying the acute phase of the SARS-CoV-2 infection. This knowledge might help reveal new biomarkers that can be utilized to recognize case severity early as well as to provide additional objective information on the effective formation of SARS-CoV-2-specific immunity and predict long-term complications of COVID-19, including a large variety of symptoms termed the 'post-COVID-19 syndrome'.
Collapse
Affiliation(s)
- Igor Kudryavtsev
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Artem Rubinstein
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Alexey Golovkin
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.G.); (O.K.)
| | - Olga Kalinina
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.G.); (O.K.)
| | - Kirill Vasilyev
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Larisa Rudenko
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Irina Isakova-Sivak
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| |
Collapse
|
32
|
Garcia-Gasalla M, Berman-Riu M, Pons J, Rodríguez A, Iglesias A, Martínez-Pomar N, Llompart-Alabern I, Riera M, Ferré Beltrán A, Figueras-Castilla A, Murillas J, Ferrer JM. Hyperinflammatory State and Low T1 Adaptive Immune Response in Severe and Critical Acute COVID-19 Patients. Front Med (Lausanne) 2022; 9:828678. [PMID: 35425776 PMCID: PMC9002349 DOI: 10.3389/fmed.2022.828678] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/07/2022] [Indexed: 01/08/2023] Open
Abstract
Background A better understanding of COVID-19 immunopathology is needed to identify the most vulnerable patients and improve treatment options. Objective We aimed to identify immune system cell populations, cytokines, and inflammatory markers related to severity in COVID-19. Methods 139 hospitalized patients with COVID-19-58 mild/moderate and 81 severe/critical-and 74 recovered patients were included in a prospective longitudinal study. Clinical data and blood samples were obtained on admission for laboratory markers, cytokines, and lymphocyte subsets study. In the recovered patients, lymphocyte subsets were analyzed 8-12 weeks after discharge. Results A National Early Warning Score 2 >2 (OR:41.4; CI:10.38-167.0), ferritin >583 pg/mL (OR:16.3; CI: 3.88-69.9), neutrophil/lymphocyte ratio >3 (OR: 3.5; CI: 1.08-12.0), sIL-2rα (sCD25) >512 pg/mL (OR: 3.3; CI: 1.48-7.9), IL-1Ra >94 pg/mL (OR: 3.2; IC: 1.4-7.3), and IL-18 >125 pg/mL (OR: 2.4; CI: 1.1-5.0) were associated with severe/critical COVID-19 in the multivariate models used. Lower absolute values of CD3, CD4, CD8, and CD19 lymphocytes together with higher frequencies of NK cells, a CD4 and CD8 activated (CD38+HLA-DR+) memory T cell and effector memory CD45RA+ (EMRA) phenotype, and lower T regulatory cell frequencies were found in severe/critical patients relative to mild/moderate and recovered COVID-19 patients. A significant reduction in Th1, Tfh1, and Tc1 with higher Th2, Tfh2, Tc2, and plasma cell frequencies was found in the most severe cases. Conclusion A characteristic hyperinflammatory state with significantly elevated neutrophil/lymphocyte ratio and ferritin, IL-1Ra, sIL-2rα, and IL-18 levels together with a "low T1 lymphocyte signature" was found in severe/critical COVID-19 patients.
Collapse
Affiliation(s)
- Mercedes Garcia-Gasalla
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - María Berman-Riu
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Jaime Pons
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Centro de Investigación Biomedica en Red (CIBER) de Enfermedades Respiratorias, Hospital Universitari Son Espases, Palma, Spain
| | - Adrián Rodríguez
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Department of Internal Medicine, Hospital Universitari Son Llàtzer, Palma, Spain
| | - Amanda Iglesias
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Centro de Investigación Biomedica en Red (CIBER) de Enfermedades Respiratorias, Hospital Universitari Son Espases, Palma, Spain
| | - Natalia Martínez-Pomar
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
| | - Isabel Llompart-Alabern
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Análisis Clínicos, Hospital Universitari Son Espases, Palma, Spain
| | - Melchor Riera
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Adrián Ferré Beltrán
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain
| | | | - Javier Murillas
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Joana M. Ferrer
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Centro de Investigación Biomedica en Red (CIBER) de Enfermedades Respiratorias, Hospital Universitari Son Espases, Palma, Spain
| |
Collapse
|
33
|
Adli A, Rahimi M, Khodaie R, Hashemzaei N, Hosseini SM. Role of Genetic Variants and Host Polymorphisms on COVID‐19: From Viral Entrance Mechanisms to Immunological Reactions. J Med Virol 2022; 94:1846-1865. [PMID: 35076118 PMCID: PMC9015257 DOI: 10.1002/jmv.27615] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/04/2022] [Accepted: 01/18/2022] [Indexed: 11/24/2022]
Abstract
Coronavirus disease 2019 (COVID‐19), caused by a highly pathogenic emerging virus, is called severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2). Knowledge regarding the pathogenesis of this virus is in infancy; however, investigation on the pathogenic mechanisms of the SARS‐CoV‐2 is underway. In COVID‐19, one of the most remarkable characteristics is the wide range of disease manifestation and severity seen across individuals of different ethnic backgrounds and geographical locations. To effectively manage COVID‐19 in the populations, beyond SARS‐CoV‐2 detection, serological response assessment, and analytic techniques, it is critical to obtain knowledge about at‐risk individuals and comprehend the identified variations in the disease's severity in general and also in the populations' levels. Several factors can contribute to variation in disease presentation, including population density, gender and age differences, and comorbid circumstances including diabetes mellitus, hypertension, and obesity. Genetic factors presumably influence SARS‐CoV‐2 infection susceptibility. Besides this, COVID‐19 has also been linked with a higher risk of mortality in men and certain ethnic groups, revealing that host genetic characteristics may affect the individual risk of death. Also, genetic variants involved in pathologic processes, including virus entrance into cells, antiviral immunity, and inflammatory response, are not entirely understood. Regarding SARS‐CoV‐2 infection characteristics, the present review suggests that various genetic polymorphisms influence virus pathogenicity and host immunity, which might have significant implications for understanding and interpreting the matter of genetics in SARS‐CoV‐2 pathogenicity and customized integrative medical care based on population investigation. Genetic factors presumably influence SARS‐CoV‐2 infection susceptibility. Genetic variants were involved in the pathologic processes of SARS‐CoV‐2 infection. Various genetic polymorphisms influence virus pathogenicity and host immunity. Human leukocyte antigens (HLAs) may play a vital role in SARS‐CoV‐2 susceptibility. Polymorphisms in several genes such as IL‐6, TMPRSS2, IFITM3, CD26, ACE, and DBP were associated with the COVID‐19 severity.
Collapse
Affiliation(s)
- Abolfazl Adli
- Human Genetic Research Center, Baqiyatallah University of Medical SciencesTehran1435916471Iran
| | - Mandana Rahimi
- Department of Pathology, School of Medicine, Hasheminejad Kidney Center, Iran University of Medical SciencesTehranIran
| | - Reza Khodaie
- Department of Biology, East Tehran Branch, Islamic Azad UniversityTehranIran
| | | | - Sayed Mostafa Hosseini
- Human Genetic Research Center, Baqiyatallah University of Medical SciencesTehran1435916471Iran
| |
Collapse
|
34
|
Abstract
Adaptive immune responses play critical roles in viral clearance and protection against re-infection, and SARS-CoV-2 is no exception. What is exceptional is the rapid characterization of the immune response to the virus performed by researchers during the first 20 months of the pandemic. This has given us a more detailed understanding of SARS-CoV-2 compared to many viruses that have been with us for a long time. Furthermore, effective COVID-19 vaccines were developed in record time, and their rollout worldwide is already making a significant difference, although major challenges remain in terms of equal access. The pandemic has engaged scientists and the public alike, and terms such as seroprevalence, neutralizing antibodies, antibody escape and vaccine certificates have become familiar to a broad community. Here, we review key findings concerning B cell and antibody (Ab) responses to SARS-CoV-2, focusing on non-severe cases and anti-spike (S) Ab responses in particular, the latter being central to protective immunity induced by infection or vaccination. The emergence of viral variants that have acquired mutations in S acutely highlights the need for continued characterization of both emerging variants and Ab responses against these during the evolving pathogen-immune system arms race.
Collapse
Affiliation(s)
- Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Ols
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Karin Loré
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | |
Collapse
|
35
|
Reyes RA, Clarke K, Gonzales SJ, Cantwell AM, Garza R, Catano G, Tragus RE, Patterson TF, Bol S, Bunnik EM. SARS-CoV-2 spike-specific memory B cells express higher levels of T-bet and FcRL5 after non-severe COVID-19 as compared to severe disease. PLoS One 2021; 16:e0261656. [PMID: 34936684 PMCID: PMC8694470 DOI: 10.1371/journal.pone.0261656] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/07/2021] [Indexed: 11/19/2022] Open
Abstract
SARS-CoV-2 infection elicits a robust B cell response, resulting in the generation of long-lived plasma cells and memory B cells. Here, we aimed to determine the effect of COVID-19 severity on the memory B cell response and characterize changes in the memory B cell compartment between recovery and five months post-symptom onset. Using high-parameter spectral flow cytometry, we analyzed the phenotype of memory B cells with reactivity against the SARS-CoV-2 spike protein or the spike receptor binding domain (RBD) in recovered individuals who had been hospitalized with non-severe (n = 8) or severe (n = 5) COVID-19. One month after symptom onset, a substantial proportion of spike-specific IgG+ B cells showed an activated phenotype. In individuals who experienced non-severe disease, spike-specific IgG+ B cells showed increased expression of markers associated with durable B cell memory, including T-bet and FcRL5, as compared to individuals who experienced severe disease. While the frequency of T-bet+ spike-specific IgG+ B cells differed between the two groups, these cells predominantly showed an activated switched memory B cell phenotype in both groups. Five months post-symptom onset, the majority of spike-specific memory B cells had a resting phenotype and the percentage of spike-specific T-bet+ IgG+ memory B cells decreased to baseline levels. Collectively, our results highlight subtle differences in the B cells response after non-severe and severe COVID-19 and suggest that the memory B cell response elicited during non-severe COVID-19 may be of higher quality than the response after severe disease.
Collapse
Affiliation(s)
- Raphael A. Reyes
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Kathleen Clarke
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - S. Jake Gonzales
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Angelene M. Cantwell
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Rolando Garza
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Gabriel Catano
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, University Health System, San Antonio, Texas, United States of America
| | - Robin E. Tragus
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, University Health System, San Antonio, Texas, United States of America
| | - Thomas F. Patterson
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, University Health System, San Antonio, Texas, United States of America
- The South Texas Veterans Health Care System, San Antonio, Texas, United States of America
| | - Sebastiaan Bol
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Evelien M. Bunnik
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
36
|
Sen K, Datta S, Ghosh A, Jha A, Ahad A, Chatterjee S, Suranjika S, Sengupta S, Bhattacharya G, Shriwas O, Avula K, Kshatri J, Prasad P, Swain R, Parida AK, Raghav SK. Single-Cell Immunogenomic Approach Identified SARS-CoV-2 Protective Immune Signatures in Asymptomatic Direct Contacts of COVID-19 Cases. Front Immunol 2021; 12:733539. [PMID: 34899693 PMCID: PMC8660575 DOI: 10.3389/fimmu.2021.733539] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/26/2021] [Indexed: 12/20/2022] Open
Abstract
The response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is largely impacted by the level of virus exposure and status of the host immunity. The nature of protection shown by direct asymptomatic contacts of coronavirus disease 2019 (COVID-19)-positive patients is quite intriguing. In this study, we have characterized the antibody titer, SARS-CoV-2 surrogate virus neutralization, cytokine levels, single-cell T-cell receptor (TCR), and B-cell receptor (BCR) profiling in asymptomatic direct contacts, infected cases, and controls. We observed significant increase in antibodies with neutralizing amplitude in asymptomatic contacts along with cytokines such as Eotaxin, granulocyte-colony stimulating factor (G-CSF), interleukin 7 (IL-7), migration inhibitory factor (MIF), and macrophage inflammatory protein-1α (MIP-1α). Upon single-cell RNA (scRNA) sequencing, we explored the dynamics of the adaptive immune response in few representative asymptomatic close contacts and COVID-19-infected patients. We reported direct asymptomatic contacts to have decreased CD4+ naive T cells with concomitant increase in CD4+ memory and CD8+ Temra cells along with expanded clonotypes compared to infected patients. Noticeable proportions of class switched memory B cells were also observed in them. Overall, these findings gave an insight into the nature of protection in asymptomatic contacts.
Collapse
Affiliation(s)
- Kaushik Sen
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India.,Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Sudeshna Datta
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India
| | - Arup Ghosh
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India.,School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Atimukta Jha
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India.,Manipal Academy of Higher Education, Manipal, India
| | - Abdul Ahad
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India.,Manipal Academy of Higher Education, Manipal, India
| | - Sanchari Chatterjee
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India.,Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Sandhya Suranjika
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India.,School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Soumya Sengupta
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India.,Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Gargee Bhattacharya
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India.,Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Omprakash Shriwas
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India.,Manipal Academy of Higher Education, Manipal, India
| | - Kiran Avula
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India.,Regional Centre for Biotechnology (RCB), Faridabad, India
| | | | - Punit Prasad
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India
| | - Rajeeb Swain
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India
| | - Ajay K Parida
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India
| | - Sunil K Raghav
- Department of Infectious Disease Biology, Institute of Life Sciences (ILS), Bhubaneswar, India.,Regional Centre for Biotechnology (RCB), Faridabad, India.,School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| |
Collapse
|
37
|
Mihaescu G, Chifiriuc MC, Vrancianu CO, Constantin M, Filip R, Popescu MR, Burlibasa L, Nicoara AC, Bolocan A, Iliescu C, Gradisteanu Pircalabioru G. Antiviral Immunity in SARS-CoV-2 Infection: From Protective to Deleterious Responses. Microorganisms 2021; 9:2578. [PMID: 34946179 PMCID: PMC8703918 DOI: 10.3390/microorganisms9122578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/26/2022] Open
Abstract
After two previous episodes, in 2002 and 2012, when two highly pathogenic coronaviruses (SARS, MERS) with a zoonotic origin emerged in humans and caused fatal respiratory illness, we are today experiencing the COVID-19 pandemic produced by SARS-CoV-2. The main question of the year 2021 is if naturally- or artificially-acquired active immunity will be effective against the evolving SARS-CoV-2 variants. This review starts with the presentation of the two compartments of antiviral immunity-humoral and cellular, innate and adaptive-underlining how the involved cellular and molecular actors are intrinsically connected in the development of the immune response in SARS-CoV-2 infection. Then, the SARS-CoV-2 immunopathology, as well as the derived diagnosis and therapeutic approaches, will be discussed.
Collapse
Affiliation(s)
- Grigore Mihaescu
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania; (G.M.); (C.O.V.); (L.B.)
| | - Mariana Carmen Chifiriuc
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania; (G.M.); (C.O.V.); (L.B.)
- Life, Environmental and Earth Sciences Division, Research Institute of the University of Bucharest, 050096 Bucharest, Romania;
- The Romanian Academy, 25 Calea Victoriei, Sector 1, 010071 Bucharest, Romania
| | | | | | - Roxana Filip
- Faculty of Medicine and Biological Sciences, Stefan cel Mare University of Suceava, 720229 Suceava, Romania;
- Regional County Emergency Hospital, 720284 Suceava, Romania
| | - Mihaela Roxana Popescu
- Department of Cardiology, Elias Emergency University Hospital “Carol Davila”, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
| | - Liliana Burlibasa
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania; (G.M.); (C.O.V.); (L.B.)
| | - Anca Cecilia Nicoara
- Faculty of Pharmacy, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
| | - Alexandra Bolocan
- General Surgery, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
| | - Ciprian Iliescu
- National Institute for Research and Development in Microtechnologies—IMT, 077190 Bucharest, Romania;
- Faculty of Applied Chemistry and Materials Science, University “Politehnica” of Bucharest, 011061 Bucharest, Romania
- Academy of Romanian Scientists, 010071 Bucharest, Romania
| | | |
Collapse
|
38
|
Does infection with or vaccination against SARS-CoV-2 lead to lasting immunity? THE LANCET. RESPIRATORY MEDICINE 2021; 9:1450-1466. [PMID: 34688434 PMCID: PMC8530467 DOI: 10.1016/s2213-2600(21)00407-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/26/2021] [Accepted: 08/21/2021] [Indexed: 12/17/2022]
Abstract
Many nations are pursuing the rollout of SARS-CoV-2 vaccines as an exit strategy from unprecedented COVID-19-related restrictions. However, the success of this strategy relies critically on the duration of protective immunity resulting from both natural infection and vaccination. SARS-CoV-2 infection elicits an adaptive immune response against a large breadth of viral epitopes, although the duration of the response varies with age and disease severity. Current evidence from case studies and large observational studies suggests that, consistent with research on other common respiratory viruses, a protective immunological response lasts for approximately 5-12 months from primary infection, with reinfection being more likely given an insufficiently robust primary humoral response. Markers of humoral and cell-mediated immune memory can persist over many months, and might help to mitigate against severe disease upon reinfection. Emerging data, including evidence of breakthrough infections, suggest that vaccine effectiveness might be reduced significantly against emerging variants of concern, and hence secondary vaccines will need to be developed to maintain population-level protective immunity. Nonetheless, other interventions will also be required, with further outbreaks likely to occur due to antigenic drift, selective pressures for novel variants, and global population mobility.
Collapse
|
39
|
Kaplonek P, Wang C, Bartsch Y, Fischinger S, Gorman MJ, Bowman K, Kang J, Dayal D, Martin P, Nowak RP, Villani AC, Hsieh CL, Charland NC, Gonye AL, Gushterova I, Khanna HK, LaSalle TJ, Lavin-Parsons KM, Lilley BM, Lodenstein CL, Manakongtreecheep K, Margolin JD, McKaig BN, Rojas-Lopez M, Russo BC, Sharma N, Tantivit J, Thomas MF, Sade-Feldman M, Feldman J, Julg B, Nilles EJ, Musk ER, Menon AS, Fischer ES, McLellan JS, Schmidt A, Goldberg MB, Filbin MR, Hacohen N, Lauffenburger DA, Alter G. Early cross-coronavirus reactive signatures of humoral immunity against COVID-19. Sci Immunol 2021; 6:eabj2901. [PMID: 34652962 PMCID: PMC8943686 DOI: 10.1126/sciimmunol.abj2901] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/06/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022]
Abstract
The introduction of vaccines has inspired hope in the battle against SARS-CoV-2. However, the emergence of viral variants, in the absence of potent antivirals, has left the world struggling with the uncertain nature of this disease. Antibodies currently represent the strongest correlate of immunity against SARS-CoV-2, thus we profiled the earliest humoral signatures in a large cohort of acutely ill (survivors and nonsurvivors) and mild or asymptomatic individuals with COVID-19. Although a SARS-CoV-2–specific immune response evolved rapidly in survivors of COVID-19, nonsurvivors exhibited blunted and delayed humoral immune evolution, particularly with respect to S2-specific antibodies. Given the conservation of S2 across β-coronaviruses, we found that the early development of SARS-CoV-2–specific immunity occurred in tandem with preexisting common β-coronavirus OC43 humoral immunity in survivors, which was also selectively expanded in individuals that develop a paucisymptomatic infection. These data point to the importance of cross-coronavirus immunity as a correlate of protection against COVID-19.
Collapse
Affiliation(s)
| | - Chuangqi Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yannic Bartsch
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | | | - Kathryn Bowman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Jaewon Kang
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Diana Dayal
- Space Exploration Technologies Corporation, Hawthorne, CA, USA
| | - Patrick Martin
- Space Exploration Technologies Corporation, Hawthorne, CA, USA
| | - Radoslaw P. Nowak
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Alexandra-Chloé Villani
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ching-Lin Hsieh
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Nicole C. Charland
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Anna L.K. Gonye
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Irena Gushterova
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Hargun K. Khanna
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Thomas J. LaSalle
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Brendan M. Lilley
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Carl L. Lodenstein
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kasidet Manakongtreecheep
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Justin D. Margolin
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Brenna N. McKaig
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Maricarmen Rojas-Lopez
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Brian C. Russo
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Nihaarika Sharma
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica Tantivit
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Molly F. Thomas
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Moshe Sade-Feldman
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jared Feldman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Boris Julg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Elon R. Musk
- Space Exploration Technologies Corporation, Hawthorne, CA, USA
| | - Anil S. Menon
- Space Exploration Technologies Corporation, Hawthorne, CA, USA
| | - Eric S. Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Aaron Schmidt
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Marcia B. Goldberg
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Michael R. Filbin
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Nir Hacohen
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| |
Collapse
|
40
|
Reyes RA, Clarke K, Gonzales SJ, Cantwell AM, Garza R, Catano G, Tragus RE, Patterson TF, Bol S, Bunnik EM. SARS-CoV-2 spike-specific memory B cells express markers of durable immunity after non-severe COVID-19 but not after severe disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.09.24.461732. [PMID: 34611662 PMCID: PMC8491845 DOI: 10.1101/2021.09.24.461732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
SARS-CoV-2 infection elicits a robust B cell response, resulting in the generation of long-lived plasma cells and memory B cells. Here, we aimed to determine the effect of COVID-19 severity on the memory B cell response and characterize changes in the memory B cell compartment between recovery and five months post-symptom onset. Using high-parameter spectral flow cytometry, we analyzed the phenotype of memory B cells with reactivity against the SARS-CoV-2 spike protein or the spike receptor binding domain (RBD) in recovered individuals who had been hospitalized with non-severe (n=8) or severe (n=5) COVID-19. One month after symptom onset, a substantial proportion of spike-specific IgG + B cells showed an activated phenotype. In individuals who experienced non-severe disease, spike-specific IgG + B cells showed increased expression of markers associated with durable B cell memory, including T-bet, FcRL5, and CD11c, which was not observed after severe disease. Five months post-symptom onset, the majority of spike-specific memory B cells had a resting phenotype and the percentage of spike-specific T-bet + IgG + memory B cells decreased to baseline levels. Collectively, our results suggest that the memory B cell response elicited during non-severe COVID-19 may be of higher quality than the response after severe disease.
Collapse
Affiliation(s)
- Raphael A. Reyes
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kathleen Clarke
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - S. Jake Gonzales
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Angelene M. Cantwell
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Rolando Garza
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Gabriel Catano
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, University Health System, San Antonio, TX, USA
| | - Robin E. Tragus
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, University Health System, San Antonio, TX, USA
| | - Thomas F. Patterson
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, University Health System, San Antonio, TX, USA
| | - Sebastiaan Bol
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Evelien M. Bunnik
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
41
|
Ligotti ME, Pojero F, Accardi G, Aiello A, Caruso C, Duro G, Candore G. Immunopathology and Immunosenescence, the Immunological Key Words of Severe COVID-19. Is There a Role for Stem Cell Transplantation? Front Cell Dev Biol 2021; 9:725606. [PMID: 34595175 PMCID: PMC8477205 DOI: 10.3389/fcell.2021.725606] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/06/2021] [Indexed: 01/08/2023] Open
Abstract
The outcomes of Coronavirus disease-2019 (COVID-19) vary depending on the age, health status and sex of an individual, ranging from asymptomatic to lethal. From an immunologic viewpoint, the final severe lung damage observed in COVID-19 should be caused by cytokine storm, driven mainly by interleukin-6 and other pro-inflammatory cytokines. However, which immunopathogenic status precedes this "cytokine storm" and why the male older population is more severely affected, are currently unanswered questions. The aging of the immune system, i.e., immunosenescence, closely associated with a low-grade inflammatory status called "inflammageing," should play a key role. The remodeling of both innate and adaptive immune response observed with aging can partly explain the age gradient in severity and mortality of COVID-19. This review discusses how aging impacts the immune response to the virus, focusing on possible strategies to rejuvenate the immune system with stem cell-based therapies. Indeed, due to immunomodulatory and anti-inflammatory properties, multipotent mesenchymal stem cells (MSCs) are a worth-considering option against COVID-19 adverse outcomes.
Collapse
Affiliation(s)
- Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Institute for Biomedical Research and Innovation, National Research Council of Italy, Palermo, Italy
| | - Fanny Pojero
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- International Society on Aging and Disease, Fort Worth, TX, United States
| | - Giovanni Duro
- Institute for Biomedical Research and Innovation, National Research Council of Italy, Palermo, Italy
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
42
|
Kealy L, Good-Jacobson KL. Advances in understanding the formation and fate of B-cell memory in response to immunization or infection. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab018. [PMID: 36845573 PMCID: PMC8499879 DOI: 10.1093/oxfimm/iqab018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/06/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
Immunological memory has the potential to provide lifelong protection against recurrent infections. As such, it has been crucial to the success of vaccines. Yet, the recent pandemic has illuminated key gaps in our knowledge related to the factors influencing effective memory formation and the inability to predict the longevity of immune protection. In recent decades, researchers have acquired a number of novel and powerful tools with which to study the factors underpinning humoral memory. These tools have been used to study the B-cell fate decisions that occur within the germinal centre (GC), a site where responding B cells undergo affinity maturation and are one of the major routes for memory B cell and high-affinity long-lived plasma cell formation. The advent of single-cell sequencing technology has provided an enhanced resolution for studying fate decisions within the GC and cutting-edge techniques have enabled researchers to model this reaction with more accuracy both in vitro and in silico. Moreover, modern approaches to studying memory B cells have allowed us to gain a better appreciation for the heterogeneity and adaptability of this vital class of B cells. Together, these studies have facilitated important breakthroughs in our understanding of how these systems operate to ensure a successful immune response. In this review, we describe recent advances in the field of GC and memory B-cell biology in order to provide insight into how humoral memory is formed, as well as the potential for generating lasting immunity to novel pathogens such as severe acute respiratory syndrome coronavirus 2.
Collapse
Affiliation(s)
- Liam Kealy
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Kim L Good-Jacobson
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia,Correspondence address. Department of Biochemistry and Molecular Biology, Monash University, Ground floor reception, 23 Innovation Walk (Bldg 77), Clayton, Victoria 3800 Australia. Tel: (+613) 990-29510; E-mail: ; Twitter: @KimLJacobson
| |
Collapse
|
43
|
Hasenkrug KJ, Feldmann F, Myers L, Santiago ML, Guo K, Barrett BS, Mickens KL, Carmody A, Okumura A, Rao D, Collins MM, Messer RJ, Lovaglio J, Shaia C, Rosenke R, van Doremalen N, Clancy C, Saturday G, Hanley P, Smith BJ, Meade-White K, Shupert WL, Hawman DW, Feldmann H. Recovery from Acute SARS-CoV-2 Infection and Development of Anamnestic Immune Responses in T Cell-Depleted Rhesus Macaques. mBio 2021; 12:e0150321. [PMID: 34311582 PMCID: PMC8406331 DOI: 10.1128/mbio.01503-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022] Open
Abstract
Severe coronavirus disease 2019 (COVID-19) has been associated with T cell lymphopenia, but no causal effect of T cell deficiency on disease severity has been established. To investigate the specific role of T cells in recovery from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, we studied rhesus macaques that were depleted of either CD4+, CD8+, or both T cell subsets prior to infection. Peak virus loads were similar in all groups, but the resolution of virus in the T cell-depleted animals was slightly delayed compared to that in controls. The T cell-depleted groups developed virus-neutralizing antibody responses and class switched to IgG. When reinfected 6 weeks later, the T cell-depleted animals showed anamnestic immune responses characterized by rapid induction of high-titer virus-neutralizing antibodies, faster control of virus loads, and reduced clinical signs. These results indicate that while T cells play a role in the recovery of rhesus macaques from acute SARS-CoV-2 infections, their depletion does not induce severe disease, and T cells do not account for the natural resistance of rhesus macaques to severe COVID-19. Neither primed CD4+ nor CD8+ T cells appeared critical for immunoglobulin class switching, the development of immunological memory, or protection from a second infection. IMPORTANCE Patients with severe COVID-19 often have decreased numbers of T cells, a cell type important in fighting most viral infections. However, it is not known whether the loss of T cells contributes to severe COVID-19 or is a consequence of it. We studied rhesus macaques, which develop only mild COVID-19, similar to most humans. Experimental depletion of T cells slightly prolonged their clearance of virus, but there was no increase in disease severity. Furthermore, they were able to develop protection from a second infection and produced antibodies capable of neutralizing the virus. They also developed immunological memory, which allows a much stronger and more rapid response upon a second infection. These results suggest that T cells are not critical for recovery from acute SARS-CoV-2 infections in this model and point toward B cell responses and antibodies as the essential mediators of protection from re-exposure.
Collapse
Affiliation(s)
- Kim J. Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Lara Myers
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Mario L. Santiago
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kejun Guo
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Bradley S. Barrett
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kaylee L. Mickens
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Aaron Carmody
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Atsushi Okumura
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Deepashri Rao
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Madison M. Collins
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Ronald J. Messer
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Rebecca Rosenke
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Neeltje van Doremalen
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Chad Clancy
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Patrick Hanley
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Brian J. Smith
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - W. Lesley Shupert
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - David W. Hawman
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
44
|
Maecker HT. Immune profiling of COVID-19: preliminary findings and implications for the pandemic. J Immunother Cancer 2021; 9:jitc-2021-002550. [PMID: 33963016 PMCID: PMC8108128 DOI: 10.1136/jitc-2021-002550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
SARS-CoV-2 infection can have widely diverse clinical outcomes, from asymptomatic infection to death, with many possible clinical symptoms and syndromes. It is thus essential to understand how the virus interacts with the host immune system to bring about these varied outcomes and to inform vaccine development. We now know that both antibody and T cell responses are induced in the majority of infected individuals, and that cross-reactive responses from other coronaviruses also exist in the uninfected population. Innate immune responses are a key focus of research and may influence the course of disease and the character of subsequent adaptive responses. Finally, baseline immune profiles and changes during early acute infection may be key to predicting the course of disease. Understanding all these aspects can help to create better immune monitoring tools for COVID-19, including tools for predicting disease severity or specific sequelae, perhaps even prior to infection.
Collapse
Affiliation(s)
- Holden T Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
45
|
Persistent COVID-19 Symptoms Minimally Impact the Development of SARS-CoV-2-Specific T Cell Immunity. Viruses 2021; 13:v13050916. [PMID: 34063463 PMCID: PMC8155927 DOI: 10.3390/v13050916] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 represents an unprecedented public health challenge. While the majority of SARS-CoV-2-infected individuals with mild-to-moderate COVID-19 resolve their infection with few complications, some individuals experience prolonged symptoms lasting for weeks after initial diagnosis. Persistent viral infections are commonly accompanied by immunologic dysregulation, but it is unclear if persistent COVID-19 impacts the development of virus-specific cellular immunity. To this end, we analyzed SARS-CoV-2-specific cellular immunity in convalescent COVID-19 patients who experienced eight days or fewer of COVID-19 symptoms or symptoms persisting for 18 days or more. We observed that persistent COVID-19 symptoms were not associated with the development of an overtly dysregulated cellular immune response. Furthermore, we observed that reactivity against the N protein from SARS-CoV-2 correlates with the amount of reactivity against the seasonal human coronaviruses 229E and NL63. These results provide insight into the processes that regulate the development of cellular immunity against SARS-CoV-2 and related human coronaviruses.
Collapse
|
46
|
Haslbauer JD, Matter MS, Stalder AK, Tzankov A. Histomorphological patterns of regional lymph nodes in COVID-19 lungs. DER PATHOLOGE 2021; 42:89-97. [PMID: 33950285 PMCID: PMC8098637 DOI: 10.1007/s00292-021-00945-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND A dysregulated immune response is considered one of the major factors leading to severe COVID-19. Previously described mechanisms include the development of a cytokine storm, missing immunoglobulin class switch, antibody-mediated enhancement, and aberrant antigen presentation. OBJECTIVES To understand the heterogeneity of immune response in COVID-19, a thorough investigation of histomorphological patterns in regional lymph nodes was performed. MATERIALS AND METHODS Lymph nodes from the cervical, mediastinal, and hilar regions were extracted from autopsies of patients with lethal COVID-19 (n = 20). Histomorphological characteristics, SARS-CoV‑2 qRT-PCR, and gene expression profiling on common genes involved in immunologic response were analyzed. RESULTS Lymph nodes displayed moderate to severe capillary stasis and edema, an increased presence of extrafollicular plasmablasts, mild to moderate plasmacytosis, a dominant population of CD8+ T‑cells, and CD11c/CD68+ histiocytosis with hemophagocytic activity. Out of 20 cases, 18 presented with hypoplastic or missing germinal centers with a decrease of follicular dendritic cells and follicular T‑helper cells. A positive viral load was detected by qRT-PCR in 14 of 20 cases, yet immunohistochemistry for SARS-CoV-2 N-antigen revealed positivity in sinus histiocytes of only one case. Gene expression analysis revealed an increased expression of STAT1, CD163, granzyme B, CD8A, MZB1, and PAK1, as well as CXCL9. CONCLUSIONS Taken together, our findings imply a dysregulated immune response in lethal COVID-19. The absence/hypoplasia of germinal centers and increased presence of plasmablasts implies a transient B‑cell response, implying an impaired development of long-term immunity against SARS-CoV‑2 in such occasions.
Collapse
Affiliation(s)
- Jasmin D Haslbauer
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland.,Institute of Medical Genetics and Pathology, Schönbeinstraße 40, 4031, Basel, Switzerland
| | - Matthias S Matter
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Anna K Stalder
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|
47
|
Hasenkrug KJ, Feldmann F, Myers L, Santiago ML, Guo K, Barrett BS, Mickens KL, Carmody A, Okumura A, Rao D, Collins MM, Messer RJ, Lovaglio J, Shaia C, Rosenke R, van Doremalen N, Clancy C, Saturday G, Hanley P, Smith B, Meade-White K, Shupert WL, Hawman DW, Feldmann H. Recovery from acute SARS-CoV-2 infection and development of anamnestic immune responses in T cell-depleted rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.04.02.438262. [PMID: 33821272 PMCID: PMC8020972 DOI: 10.1101/2021.04.02.438262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2023]
Abstract
Severe COVID-19 has been associated with T cell lymphopenia 1,2, but no causal effect of T cell deficiency on disease severity has been established. To investigate the specific role of T cells in recovery from SARS-CoV-2 infections we studied rhesus macaques that were depleted of either CD4+, CD8+ or both T cell subsets prior to infection. Peak virus loads were similar in all groups, but the resolution of virus in the T cell-depleted animals was slightly delayed compared to controls. The T cell-depleted groups developed virus-neutralizing antibody responses and also class-switched to IgG. When re-infected six weeks later, the T cell-depleted animals showed anamnestic immune responses characterized by rapid induction of high-titer virus-neutralizing antibodies, faster control of virus loads and reduced clinical signs. These results indicate that while T cells play a role in the recovery of rhesus macaques from acute SARS-CoV-2 infections, their depletion does not induce severe disease, and T cells do not account for the natural resistance of rhesus macaques to severe COVID-19. Neither primed CD4+ or CD8+ T cells appeared critical for immunoglobulin class switching, the development of immunological memory or protection from a second infection.
Collapse
Affiliation(s)
- Kim J. Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Lara Myers
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Mario L. Santiago
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Kejun Guo
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Bradley S. Barrett
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Kaylee L. Mickens
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Aaron Carmody
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Atsushi Okumura
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Deepashri Rao
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Madison M. Collins
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Ronald J. Messer
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Rebecca Rosenke
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Neeltje van Doremalen
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Chad Clancy
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Patrick Hanley
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Brian Smith
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - W. Lesley Shupert
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - David W. Hawman
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
48
|
Sagiv E, Portman MA. CD24 for Cardiovascular Researchers: A Key Molecule in Cardiac Immunology, Marker of Stem Cells and Target for Drug Development. J Pers Med 2021; 11:jpm11040260. [PMID: 33915986 PMCID: PMC8066264 DOI: 10.3390/jpm11040260] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022] Open
Abstract
The study of the membrane protein, CD24, and its emerging role in major disease processes, has made a huge leap forward in the past two decades. It appears to have various key roles in oncogenesis, tumor progression and metastasis, stem cell maintenance and immune modulation. First described in the 1980s as the homologous human protein to the mouse HSA (Heat Stable Antigen), it was reported as a surface marker in developing hematopoietic cell lines. The later discovery of its overexpression in a large number of human neoplasms, lead cancer researchers to discover its various active roles in critical checkpoints during cancer development and progression. Targeting CD24 in directed drug development showed promising results in cancer treatment. More recently, the chimeric CD24-Fc protein has shown exciting results in clinical trials as a specific modulator of auto-inflammatory syndromes. This report is aimed to summarize the relevant literature on CD24 and tie it together with recent advancements in cardiovascular research. We hypothesize that CD24 is a promising focus of research in the understanding of cardiovascular disease processes and the development of novel biological therapies.
Collapse
Affiliation(s)
- Eyal Sagiv
- Correspondence: ; Tel.: +1-206-987-6916; Fax: +1-206-987-3839
| | | |
Collapse
|
49
|
Gasser R, Cloutier M, Prévost J, Fink C, Ducas É, Ding S, Dussault N, Landry P, Tremblay T, Laforce-Lavoie A, Lewin A, Beaudoin-Bussières G, Laumaea A, Medjahed H, Larochelle C, Richard J, Dekaban GA, Dikeakos JD, Bazin R, Finzi A. Major role of IgM in the neutralizing activity of convalescent plasma against SARS-CoV-2. Cell Rep 2021; 34:108790. [PMID: 33596407 PMCID: PMC7874916 DOI: 10.1016/j.celrep.2021.108790] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/05/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
Characterization of the humoral response to SARS-CoV-2, the etiological agent of COVID-19, is essential to help control the infection. The neutralization activity of plasma from patients with COVID-19 decreases rapidly during the first weeks after recovery. However, the specific role of each immunoglobulin isotype in the overall neutralizing capacity is still not well understood. In this study, we select plasma from a cohort of convalescent patients with COVID-19 and selectively deplete immunoglobulin A, M, or G before testing the remaining neutralizing capacity of the depleted plasma. We find that depletion of immunoglobulin M is associated with the most substantial loss of virus neutralization, followed by immunoglobulin G. This observation may help design efficient antibody-based COVID-19 therapies and may also explain the increased susceptibility to SARS-CoV-2 of autoimmune patients receiving therapies that impair the production of immunoglobulin M (IgM).
Collapse
Affiliation(s)
- Romain Gasser
- Centre de recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Marc Cloutier
- Héma-Québec, Affaires Médicales et Innovation, Québec, QC G1V 5C3, Canada
| | - Jérémie Prévost
- Centre de recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Corby Fink
- Biotherapeutics Research Laboratory, Robarts Research Institute, London, ON NGA 5B7, Canada; Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5B7, Canada
| | - Éric Ducas
- Héma-Québec, Affaires Médicales et Innovation, Québec, QC G1V 5C3, Canada
| | - Shilei Ding
- Centre de recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Nathalie Dussault
- Héma-Québec, Affaires Médicales et Innovation, Québec, QC G1V 5C3, Canada
| | - Patricia Landry
- Héma-Québec, Affaires Médicales et Innovation, Québec, QC G1V 5C3, Canada
| | - Tony Tremblay
- Héma-Québec, Affaires Médicales et Innovation, Québec, QC G1V 5C3, Canada
| | | | - Antoine Lewin
- Héma-Québec, Affaires Médicales et Innovation, Montréal, QC H4R 2W7, Canada; Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Guillaume Beaudoin-Bussières
- Centre de recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Annemarie Laumaea
- Centre de recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | | | - Catherine Larochelle
- Centre de recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada; Department of Neurosciences, University of Montreal, Montreal, QC H2X 0A9, Canada
| | - Jonathan Richard
- Centre de recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Gregory A Dekaban
- Biotherapeutics Research Laboratory, Robarts Research Institute, London, ON NGA 5B7, Canada; Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5B7, Canada
| | - Jimmy D Dikeakos
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5B7, Canada
| | - Renée Bazin
- Héma-Québec, Affaires Médicales et Innovation, Québec, QC G1V 5C3, Canada.
| | - Andrés Finzi
- Centre de recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada.
| |
Collapse
|
50
|
Hartley GE, Edwards ESJ, Aui PM, Varese N, Stojanovic S, McMahon J, Peleg AY, Boo I, Drummer HE, Hogarth PM, O'Hehir RE, van Zelm MC. Rapid generation of durable B cell memory to SARS-CoV-2 spike and nucleocapsid proteins in COVID-19 and convalescence. Sci Immunol 2021; 5:5/54/eabf8891. [PMID: 33443036 PMCID: PMC7877496 DOI: 10.1126/sciimmunol.abf8891] [Citation(s) in RCA: 223] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/18/2020] [Indexed: 12/13/2022]
Abstract
Lasting immunity following SARS-CoV-2 infection is questioned because serum antibodies decline in convalescence. However, functional immunity is mediated by long-lived memory T and B (Bmem) cells. Therefore, we generated fluorescently-labeled tetramers of the spike receptor binding domain (RBD) and nucleocapsid protein (NCP) to determine the longevity and immunophenotype of SARS-CoV-2-specific Bmem cells in COVID-19 patients. A total of 36 blood samples were obtained from 25 COVID-19 patients between 4 and 242 days post-symptom onset including 11 paired samples. While serum IgG to RBD and NCP was identified in all patients, antibody levels began declining at 20 days post-symptom onset. RBD- and NCP-specific Bmem cells predominantly expressed IgM+ or IgG1+ and continued to rise until 150 days. RBD-specific IgG+ Bmem were predominantly CD27+, and numbers significantly correlated with circulating follicular helper T cell numbers. Thus, the SARS-CoV-2 antibody response contracts in convalescence with persistence of RBD- and NCP-specific Bmem cells. Flow cytometric detection of SARS-CoV-2-specific Bmem cells enables detection of long-term immune memory following infection or vaccination for COVID-19.
Collapse
Affiliation(s)
- Gemma E Hartley
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Emily S J Edwards
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Pei M Aui
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Nirupama Varese
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,Department of Allergy, Immunology & Respiratory Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Stephanie Stojanovic
- Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, VIC, Australia
| | - James McMahon
- Department of Infectious Diseases, The Alfred and Central Clinical school, Monash University, Melbourne, VIC, Australia.,Department of Infectious Diseases, Monash Health, Melbourne, VIC, Australia
| | - Anton Y Peleg
- Department of Infectious Diseases, The Alfred and Central Clinical school, Monash University, Melbourne, VIC, Australia.,Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Irene Boo
- Viral Entry and Vaccines Group, Burnet Institute, Melbourne, VIC, Australia
| | - Heidi E Drummer
- Viral Entry and Vaccines Group, Burnet Institute, Melbourne, VIC, Australia.,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - P Mark Hogarth
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,Immune Therapies Group, Burnet Institute, Melbourne, VIC, Australia.,Department of Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Robyn E O'Hehir
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,Department of Allergy, Immunology & Respiratory Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, VIC, Australia
| | - Menno C van Zelm
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia. .,Department of Allergy, Immunology & Respiratory Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|