SARS-CoV-2 spike antigen-specific B cell and antibody responses in pre-vaccination period COVID-19 convalescent males and females with or without post-covid condition

Background Following SARS-CoV-2 infection a significant proportion of convalescent individuals develop the post-COVID condition (PCC) that is characterized by wide spectrum of symptoms encompassing various organs. Even though the underlying pathophysiology of PCC is not known, detection of viral transcripts and antigens in tissues other than lungs raise the possibility that PCC may be a consequence of aberrant immune response to the viral antigens. To test this hypothesis, we evaluated B cell and antibody responses to the SARS-CoV-2 antigens in PCC patients who experienced mild COVID-19 disease during the pre-vaccination period of COVID-19 pandemic. Methods The study subjects included unvaccinated male and female subjects who developed PCC or not (No-PCC) after clearing RT-PCR confirmed mild COVID-19 infection. SARS-CoV-2 D614G and omicron RBD specific B cell subsets in peripheral circulation were assessed by flow cytometry. IgG, IgG3 and IgA antibody titers toward RBD, spike and nucleocapsid antigens in the plasma were evaluated by ELISA. Results The frequency of the B cells specific to D614G-RBD were comparable in convalescent groups with and without PCC in both males and females. Notably, in females with PCC, the anti-D614G RBD specific double negative (IgD-CD27-) B cells showed significant correlation with the number of symptoms at acute of infection. Anti-spike antibody responses were also higher at 3 months post-infection in females who developed PCC, but not in the male PCC group. On the other hand, the male PCC group also showed consistently high anti-RBD IgG responses compared to all other groups. Conclusions The antibody responses to the spike protein, but not the RBD-specific B cell responses diverge between convalescent males and females, and those who develop PCC or not. Our findings suggest that sex-related factors may also be involved in the development of PCC via modulating antibody responses to the SARS-CoV-2 antigens. Short Summary Post-COVID Condition (PCC) is lingering illness that afflicts a significant proportion of COVID-19 patients from three months after clearing SARS-CoV-2 infection. Therapy for PCC is only palliative and the underlying disease mechanisms are unclear. The wide spectrum of PCC symptoms that can affect different organs and the detection of viral components in tissues distant from lungs raise the possibility that PCC may be associated with aberrant immune response due to presence of viral antigens. Therefore, we studied B cell and antibody responses to the spike and nucleoprotein antigens in PCC patients who cleared mild SARS-CoV-2 infection during the pre-vaccination COVID-19 pandemic period. We observed divergent patterns of immune reactivity to the spike protein in PCC males and females at different times post-infection, suggesting that the immune responses in PCC may also be influenced by sex-related factors.

[1]  J. Pedrosa,et al.  Post-acute sequelae of COVID-19 is characterized by diminished peripheral CD8+β7 integrin+ T cells and anti-SARS-CoV-2 IgA response , 2023, Nature Communications.

[2]  J. Fraussen,et al.  IgD-CD27- double negative (DN) B cells: origins and functions in health and disease. , 2023, Immunology letters.

[3]  C. Goldzweig,et al.  Serological response to vaccination in post-acute sequelae of COVID , 2023, BMC Infectious Diseases.

[4]  W. Lau,et al.  Influenza vaccination reveals sex dimorphic imprints of prior mild COVID-19 , 2023, Nature.

[5]  A. Piché,et al.  Prevalence of persistent symptoms at least 1 month after SARS-CoV-2 Omicron infection in adults. , 2022, Journal of the Association of Medical Microbiology and Infectious Disease Canada = Journal officiel de l'Association pour la microbiologie medicale et l'infectiologie Canada.

[6]  A. Piché,et al.  SARS-CoV-2 infection outcomes associated with the Delta variant: A prospective cohort study. , 2022, Journal of the Association of Medical Microbiology and Infectious Disease Canada = Journal officiel de l'Association pour la microbiologie medicale et l'infectiologie Canada.

[7]  Sarah E. Webster,et al.  Age-related changes in antigen-specific natural antibodies are influenced by sex , 2023, Frontiers in Immunology.

[8]  S. Pittaluga,et al.  SARS-CoV-2 infection and persistence in the human body and brain at autopsy , 2022, Nature.

[9]  J. Arrazola,et al.  Multimodal neuroimaging in post-COVID syndrome and correlation with cognition , 2022, Brain : a journal of neurology.

[10]  Christopher J. L. Murray,et al.  Estimated Global Proportions of Individuals With Persistent Fatigue, Cognitive, and Respiratory Symptom Clusters Following Symptomatic COVID-19 in 2020 and 2021. , 2022, JAMA.

[11]  James N. Druckman,et al.  Prevalence and Correlates of Long COVID Symptoms Among US Adults , 2022, JAMA network open.

[12]  B. Palmer,et al.  Autoantibodies elicited with SARS-CoV-2 infection are linked to alterations in double negative B cells , 2022, Frontiers in Immunology.

[13]  G. Alter,et al.  Persistent circulating SARS-CoV-2 spike is associated with post-acute COVID-19 sequelae , 2022, medRxiv.

[14]  T. Ndung’u,et al.  HIV skews the SARS-CoV-2 B cell response towards an extrafollicular maturation pathway , 2022, bioRxiv.

[15]  A. G. Boef,et al.  Sex-Related Differences in the Immune Response to Meningococcal Vaccinations During Adolescence , 2022, Frontiers in Public Health.

[16]  D. McGovern,et al.  Demographic and clinical characteristics associated with variations in antibody response to BNT162b2 COVID-19 vaccination among healthcare workers at an academic medical centre: a longitudinal cohort analysis , 2022, BMJ Open.

[17]  J. Lieberman,et al.  FcγR-mediated SARS-CoV-2 infection of monocytes activates inflammation , 2022, Nature.

[18]  J. Alcorn,et al.  SARS-CoV-2 Antibody Response Is Associated with Age and Body Mass Index in Convalescent Outpatients , 2022, The Journal of Immunology.

[19]  Paloma Martin,et al.  Post-COVID-19 syndrome. SARS-CoV-2 RNA detection in plasma, stool, and urine in patients with persistent symptoms after COVID-19 , 2022, BMC Infectious Diseases.

[20]  C. Carty,et al.  Combinatorial analysis reveals highly coordinated early-stage immune reactions that predict later antiviral immunity in mild COVID-19 patients , 2022, Cell Reports Medicine.

[21]  M. Peluso,et al.  Early clues regarding the pathogenesis of long-COVID , 2022, Trends in Immunology.

[22]  M. Puhan,et al.  Immunoglobulin signature predicts risk of post-acute COVID-19 syndrome , 2022, Nature Communications.

[23]  A. Gingras,et al.  A scalable serology solution for profiling humoral immune responses to SARS‐CoV‐2 infection and vaccination , 2022, Clinical & translational immunology.

[24]  Inyoul Y. Lee,et al.  Multiple early factors anticipate post-acute COVID-19 sequelae , 2022, Cell.

[25]  D. Vézina,et al.  Temporal associations of B and T cell immunity with robust vaccine responsiveness in a 16-week interval BNT162b2 regimen , 2021, bioRxiv.

[26]  Tuba Ozgocer,et al.  Analysis of long‐term antibody response in COVID‐19 patients by symptoms grade, gender, age, BMI, and medication , 2021, Journal of medical virology.

[27]  M. Peluso,et al.  Markers of Immune Activation and Inflammation in Individuals With Postacute Sequelae of Severe Acute Respiratory Syndrome Coronavirus 2 Infection. , 2021, The Journal of infectious diseases.

[28]  C. Prudêncio,et al.  Assessment of avidity related to IgG subclasses in SARS-CoV-2 Brazilian infected patients , 2021, Scientific Reports.

[29]  M. Peluso,et al.  Long-term SARS-CoV-2-specific immune and inflammatory responses in individuals recovering from COVID-19 with and without post-acute symptoms , 2021, Cell Reports.

[30]  B. Mustanski,et al.  COVID-19 mRNA Vaccination Generates Greater Immunoglobulin G Levels in Women Compared to Men , 2021, The Journal of infectious diseases.

[31]  V. Gant,et al.  Long COVID following mild SARS-CoV-2 infection: characteristic T cell alterations and response to antihistamines , 2021, medRxiv.

[32]  J. Tang,et al.  Rapid tests for quantification of infectiousness are urgently required in patients with COVID-19 , 2021, The Lancet Microbe.

[33]  A. Mehta,et al.  Longitudinal analysis shows durable and broad immune memory after SARS-CoV-2 infection with persisting antibody responses and memory B and T cells , 2021, medRxiv.

[34]  D. Brodie,et al.  Post-acute COVID-19 syndrome , 2021, Nature Medicine.

[35]  David A. Drew,et al.  Attributes and predictors of long COVID , 2021, Nature Medicine.

[36]  M. Cecconi,et al.  Long COVID hallmarks on [18F]FDG-PET/CT: a case-control study , 2021, European Journal of Nuclear Medicine and Molecular Imaging.

[37]  R. Tedder,et al.  The Association Between Antibody Response to Severe Acute Respiratory Syndrome Coronavirus 2 Infection and Post–COVID-19 Syndrome in Healthcare Workers , 2021, The Journal of infectious diseases.

[38]  M. Davenport,et al.  Evolution of immune responses to SARS-CoV-2 in mild-moderate COVID-19 , 2021, Nature communications.

[39]  A. Morice,et al.  Post-COVID-19 Symptom Burden: What is Long-COVID and How Should We Manage It? , 2021, Lung.

[40]  D. Raoult,et al.  18F-FDG brain PET hypometabolism in patients with long COVID , 2021, European Journal of Nuclear Medicine and Molecular Imaging.

[41]  Bjoern Peters,et al.  Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection , 2021, Science.

[42]  A. Meola,et al.  Maturation and persistence of the anti-SARS-CoV-2 memory B cell response , 2020, Cell.

[43]  A. Casto,et al.  Dynamics of Neutralizing Antibody Titers in the Months After Severe Acute Respiratory Syndrome Coronavirus 2 Infection , 2020, The Journal of infectious diseases.

[44]  M. Rattray,et al.  Sex differences in innate anti-viral immune responses to respiratory viruses and in their clinical outcomes in a birth cohort study , 2020, Scientific Reports.

[45]  A. Fontanet,et al.  Sex Differences in the Evolution of Neutralizing Antibodies to Severe Acute Respiratory Syndrome Coronavirus 2 , 2021, The Journal of Infectious Diseases.

[46]  M. Shlomchik,et al.  Germinal Center and Extrafollicular B Cell Responses in Vaccination, Immunity, and Autoimmunity. , 2020, Immunity.

[47]  H. Peckham,et al.  Male sex identified by global COVID-19 meta-analysis as a risk factor for death and ITU admission , 2020, Nature Communications.

[48]  M. Nussenzweig,et al.  Evolution of Antibody Immunity to SARS-CoV-2 , 2020, bioRxiv.

[49]  O. Laeyendecker,et al.  Healthy donor T-cell responses to common cold coronaviruses and SARS-CoV-2. , 2020, The Journal of clinical investigation.

[50]  S. Nahnsen,et al.  Specific Induction of Double Negative B Cells During Protective and Pathogenic Immune Responses , 2020, bioRxiv.

[51]  M. Veldhoen,et al.  Seroprevalence of anti‐SARS‐CoV‐2 antibodies in COVID‐19 patients and healthy volunteers up to 6 months post disease onset , 2020, medRxiv.

[52]  Kelsey K. Finn,et al.  Loss of Bcl-6-Expressing T Follicular Helper Cells and Germinal Centers in COVID-19 , 2020, Cell.

[53]  L. Carter,et al.  Functional SARS-CoV-2-specific immune memory persists after mild COVID-19 , 2020, medRxiv.

[54]  D. Raoult,et al.  18F-FDG brain PET hypometabolism in post-SARS-CoV-2 infection: substrate for persistent/delayed disorders? , 2020, European Journal of Nuclear Medicine and Molecular Imaging.

[55]  V. Martel-Laferrière,et al.  Decline of Humoral Responses against SARS-CoV-2 Spike in Convalescent Individuals , 2020, mBio.

[56]  Angelo Carfì,et al.  Persistent Symptoms in Patients After Acute COVID-19. , 2020, JAMA.

[57]  C. Rice,et al.  Convergent Antibody Responses to SARS-CoV-2 in Convalescent Individuals , 2020, Nature.

[58]  Victor G. Puelles,et al.  Multiorgan and Renal Tropism of SARS-CoV-2 , 2020, The New England journal of medicine.

[59]  W. Chen,et al.  Critically ill SARS-CoV-2 patients display lupus-like hallmarks of extrafollicular B cell activation , 2020, medRxiv.

[60]  J. McCarthy,et al.  Infection-induced plasmablasts are a nutrient sink that impairs humoral immunity to malaria , 2020, Nature Immunology.

[61]  Scott A. Jenks,et al.  Challenges and Opportunities for Consistent Classification of Human B Cell and Plasma Cell Populations , 2019, Front. Immunol..

[62]  S. Klein,et al.  The intersection of sex and gender in the treatment of influenza. , 2019, Current opinion in virology.

[63]  G. Alter,et al.  Sex differences in vaccine-induced humoral immunity , 2018, Seminars in Immunopathology.

[64]  Jie Cui,et al.  Origin and evolution of pathogenic coronaviruses , 2018, Nature Reviews Microbiology.

[65]  G. Gibson,et al.  Distinct Effector B Cells Induced by Unregulated Toll‐like Receptor 7 Contribute to Pathogenic Responses in Systemic Lupus Erythematosus , 2018, Immunity.

[66]  S. Klein,et al.  Sex differences in immune responses , 2016, Nature Reviews Immunology.

[67]  D. Kelly,et al.  Sex-dependent immune responses to infant vaccination: an individual participant data meta-analysis of antibody and memory B cells. , 2016, Vaccine.

[68]  G. Olinger,et al.  Long-term sequelae after Ebola virus disease in Bundibugyo, Uganda: a retrospective cohort study. , 2015, The Lancet. Infectious diseases.

[69]  Susan Kovats,et al.  Estrogen receptors regulate innate immune cells and signaling pathways , 2015, Cellular immunology.

[70]  L. BenMohamed,et al.  Gender-Dependent HLA-DR-Restricted Epitopes Identified from Herpes Simplex Virus Type 1 Glycoprotein D , 2008, Clinical and Vaccine Immunology.

[71]  J. McCluskey,et al.  Immunodominance Hierarchies and Gender Bias in Direct TCD8‐Cell Alloreactivity , 2007, American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons.

[72]  M. Falagas,et al.  Sex differences in the incidence and severity of respiratory tract infections. , 2007, Respiratory medicine.

[73]  I. Hickie,et al.  Post-infective and chronic fatigue syndromes precipitated by viral and non-viral pathogens: prospective cohort study , 2006, BMJ : British Medical Journal.

[74]  E. D. Kilbourne Influenza Pandemics of the 20th Century , 2006, Emerging infectious diseases.

[75]  A. Earnest,et al.  Pulmonary function and exercise capacity in survivors of Severe Acute Respiratory Syndrome , 2004, European Respiratory Journal.

[76]  G. Navis,et al.  Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis , 2004, The Journal of pathology.

[77]  K. Rajewsky,et al.  Human Immunoglobulin (Ig)M+IgD+ Peripheral Blood B Cells Expressing the CD27 Cell Surface Antigen Carry Somatically Mutated Variable Region Genes: CD27 as a General Marker for Somatically Mutated (Memory) B Cells , 1998, The Journal of experimental medicine.

[78]  L. Mitchell,et al.  Differential antibody responses to rubella virus infection in males and females. , 1992, The Journal of infectious diseases.

[79]  D. Tyrrell,et al.  The time course of the immune response to experimental coronavirus infection of man , 1990, Epidemiology and Infection.

[80]  N. Chiorazzi,et al.  Regulation of the immune response by sex hormones. I. In vitro effects of estradiol and testosterone on pokeweed mitogen-induced human B cell differentiation. , 1988, Journal of immunology.

[81]  H. Adlercreutz,et al.  Sex hormone regulation of in vitro immune response. Estradiol enhances human B cell maturation via inhibition of suppressor T cells in pokeweed mitogen-stimulated cultures , 1981, The Journal of experimental medicine.

[82]  R. Serfling,et al.  Excess pneumonia-influenza mortality by age and sex in three major influenza A2 epidemics, United States, 1957-58, 1960 and 1963. , 1967, American journal of epidemiology.