1
|
Akhtar E, Kuddusi RU, Talukder MT, Jakarea M, Haq MA, Hossain MS, Vandenent M, Islam MZ, Zaman RU, Razzaque A, Sarker P, Raqib R. Functional T cell response to COVID-19 vaccination with or without natural infection with SARS-CoV-2 in adults and children. Sci Rep 2025; 15:13341. [PMID: 40247005 PMCID: PMC12006499 DOI: 10.1038/s41598-025-95870-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
Severe COVID-19 is rare in children suggesting differences in immune response between children and adults. Limited information is available on how cellular immunity is modulated by COVID-19 vaccination and prior infection, and whether it is differentially modulated in children compared to adults. Here, we aimed to compare COVID-19 vaccine-induced functional T cell response between adults and children with and without previous SARS-CoV-2 infection. Adults (18-45 years; n = 45) and children (5-10 years; n = 51;), who received Pfizer-BioNTech COVID-19 vaccine or remained unvaccinated, and previously infected or not with SARS-CoV-2 were selected from two cross-sectional SARS-CoV-2 serosurveillance studies conducted in Bangladesh. Plasma nucleocapsid (N)-specific antibodies were measured by electrochemiluminescence immunoassay; IFN-γ, perforin and granzyme B secreting T cells were assessed using ELISpot assay. Vaccination in adults without previous infection, induced higher frequencies of IFN-γ and granzyme B secreting T lymphocytes compared to unvaccinated adults, while it increased only IFN-γ expression in vaccinated children. Previous infection increased IFN-γ response in unvaccinated adults only. Unvaccinated children showed higher granzyme B expression compared to adults irrespective of infection status. In vaccinated individuals, prior infection induced perforin expression in both adults and children. Children showed slightly different functional T cell response than adults in response to COVID-19 vaccination and infection. mRNA vaccination provided higher IFN-γ response in both adults and children, but induced cytotoxic T lymphocyte (CTL) response in adults only. Future studies may evaluate the impact of other types of COVID-19 vaccines on functional T cell immunity in children to confirm the findings.
Collapse
Affiliation(s)
- Evana Akhtar
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Rakib Ullah Kuddusi
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Tanvir Talukder
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Jakarea
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Ahsanul Haq
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Shamim Hossain
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | | | | | - Rashid U Zaman
- Foreign, Commonwealth and Development Office, British High Commission, Dhaka, 1212, Bangladesh
| | - Abdur Razzaque
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Protim Sarker
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Rubhana Raqib
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh.
| |
Collapse
|
2
|
Trimbake D, Singh D, K. YG, Babar P, S. VD, Tripathy AS. Durability of Functional SARS-CoV-2-Specific Immunological Memory and T Cell Response up to 8-9 Months Postrecovery From COVID-19. J Immunol Res 2025; 2025:9743866. [PMID: 39963186 PMCID: PMC11832264 DOI: 10.1155/jimr/9743866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025] Open
Abstract
Research on long-term follow-up in individuals who have recovered from coronavirus disease-19 (COVID-19) would yield insights regarding their immunity status and identify those who need booster vaccinations. This study evaluated the longevity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific cellular and humoral memory responses, as well as T cell effector functionalities, at 1-2 months (n = 40), 8-9 months (n = 40), and 12 months/1 year (n = 27) following recovery from SARS-CoV-2 infection. CTL response by enzyme-linked immunospot (ELISPOT); levels of cytokine by Bio-Plex, natural killer (NK), CD4+ helper, and CD8+ cytotoxic T cell functionalities using flow cytometry; anti-SARS-CoV-2 IgG by ELISA; and levels of neutralizing antibodies (NAbs) by surrogate virus NAb assay were assessed. The levels of SARS-CoV-2-specific IgG and NAb at 1-2 and 8-9 months postrecovery were hand in hand and appeared declining. SARS-CoV-2-specific B, memory B and plasma cells, and T cells sustained up to 8-9 months. Increased expression of CD107a/IFN-γ by NK cells and cytotoxic T cells at 8-9 months could be indicative of SARS-CoV-2-specific effector functions. Recovered individuals with positive and negative IgG antibody status displayed T cell response up to 1 year and 8-9 months, respectively, emphasizing the durabilty of effector immunity up to 8-9 months regardless of IgG antibody status. Overall, the recovered individuals exhibited robust immunological memory, sustained T cell response with effector functionality against SARS-CoV-2 that persists for at least 8-9 months.
Collapse
Affiliation(s)
- Diptee Trimbake
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Dharmendra Singh
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Yogesh Gurav K.
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Prasad Babar
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Varsha Dange S.
- Department of Medicine, Pimpri Chinchwad Municipal Corporation, Pimpri, Pune 411018, Maharashtra, India
| | - Anuradha S. Tripathy
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| |
Collapse
|
3
|
Pozdnyakova V, Weber B, Cheng S, Ebinger JE. Review of Immunologic Manifestations of COVID-19 Infection and Vaccination. Rheum Dis Clin North Am 2025; 51:111-121. [PMID: 39550100 DOI: 10.1016/j.rdc.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2024]
Abstract
We herein summarize currently available and clinically relevant information regarding the human immune responses to SARS-CoV-2 infection and vaccination, in relation to COVID-19 outcomes with a focus on acute respiratory distress syndrome (ARDS) and myocarditis.
Collapse
Affiliation(s)
- Valeriya Pozdnyakova
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, D4005, Los Angeles, CA 90048, USA
| | - Brittany Weber
- Carl J. and Ruth Shapiro Cardiovascular Center, Brigham and Women's Hospital, 70 Francis Street, Boston, MA 02115, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South Vicente Boulevard, Suite A3100, Los Angeles, CA 90048, USA
| | - Joseph E Ebinger
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South Vicente Boulevard, Suite A3100, Los Angeles, CA 90048, USA.
| |
Collapse
|
4
|
Paules CI, Nordwall JA, Shaw-Saliba K, Aberg JA, Gardner EM, Goodman AL, Kumarasamy N, Vasudeva S, Vock DM, North CM, Lundgren J, Aggarwal NR. Blood absolute lymphocyte count and trajectory are important in understanding severe COVID-19. BMC Infect Dis 2025; 25:67. [PMID: 39810077 PMCID: PMC11734232 DOI: 10.1186/s12879-024-10428-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Low blood absolute lymphocyte count (ALC) may predict severe COVID-19 outcomes. Knowledge gaps remain regarding the relationship of ALC trajectory with clinical outcomes and factors associated with lymphopenia. METHODS Our post hoc analysis of the Therapeutics for Inpatients with COVID-19 platform trial utilized proportional hazards models to assess relationships between Day (D) 0 lymphopenia (ALC < 0.9 cells/uL), D0 severe lymphopenia (ALC < 0.5 cells/uL) or lymphopenia trajectory between D0 and D5 with mortality and secondary infections, and with sustained recovery using Fine-Gray models. Logistic regression was used to assess relationships between clinical variables and D0 lymphopenia or lymphopenia trajectory. RESULTS D0 lymphopenia (1426/2579) and severe lymphopenia (636/2579) were associated with increased mortality (aHR 1.48; 1.08, 2.05, p = 0.016 and aHR 1.60; 1.20, 2.14, p = 0.001) and decreased recovery (aRRR 0.90; 0.82, 0.99, p = 0.033 and aRRR 0.78; 0.70, 0.87, p < 0.001 respectively). Trial participants with persistent D5 lymphopenia had increased mortality, and increased secondary infections, and participants with persistent or new lymphopenia had impaired recovery, as compared to participants with no lymphopenia. Persistent and new lymphopenia were associated with older age, male sex; prior immunosuppression, heart failure, aspirin use, and normal body mass index; biomarkers of organ damage (renal and lung), and ineffective immune response (elevated IL-6 and viral nucleocapsid antigen levels). Similar results were observed with severe lymphopenia. CONCLUSIONS Lymphopenia was predictive of severe COVID-19 outcomes, particularly when persistent or new during hospitalization. A better understanding of the underlying risk factors for lymphopenia will help illuminate disease pathogenesis and guide management strategies.
Collapse
Affiliation(s)
- Catharine I Paules
- Division of Infectious Diseases, Penn State Health Milton S. Hershey Medical Center, 500 University Drive, Hershey, PA, 17033, USA.
| | - Jacqueline A Nordwall
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Kathryn Shaw-Saliba
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Judith A Aberg
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Anna L Goodman
- MRC Clinical Trials Unit at University College London and CIDR, King's College London and Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - N Kumarasamy
- VHS Infectious Diseases Medical Centre, CART Clinical Research Site, Voluntary Health Services, Chennai, India
| | - Shikha Vasudeva
- Division of Infectious Diseases, VA Medical Center, Salem, VA, USA
| | - David M Vock
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Crystal M North
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jens Lundgren
- CHIP Center of Excellence for Health, Immunity, and Infections, Department of Infectious Diseases, University of Copenhagen, Righospitalet, Copenhagen, Denmark
| | - Neil R Aggarwal
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
5
|
Snyder TM, Gittelman RM, Klinger M, May DH, Osborne EJ, Taniguchi R, Jabran Zahid H, Kaplan IM, Dines JN, Noakes MT, Pandya R, Chen X, Elasady S, Svejnoha E, Ebert P, Pesesky MW, De Almeida P, O’Donnell H, DeGottardi Q, Keitany G, Lu J, Vong A, Elyanow R, Fields P, Al-Asadi H, Greissl J, Baldo L, Semprini S, Cerchione C, Nicolini F, Mazza M, Delmonte OM, Dobbs K, Laguna-Goya R, Carreño-Tarragona G, Barrio S, Imberti L, Sottini A, Quiros-Roldan E, Rossi C, Biondi A, Bettini LR, D’Angio M, Bonfanti P, Tompkins MF, Alba C, Dalgard C, Sambri V, Martinelli G, Goldman JD, Heath JR, Su HC, Notarangelo LD, Paz-Artal E, Martinez-Lopez J, Howie B, Carlson JM, Robins HS. Magnitude and dynamics of the T-cell response to SARS-CoV-2 infection at both individual and population levels. Front Immunol 2025; 15:1488860. [PMID: 39840037 PMCID: PMC11747429 DOI: 10.3389/fimmu.2024.1488860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/05/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction T cells are involved in the early identification and clearance of viral infections and also support the development of antibodies by B cells. This central role for T cells makes them a desirable target for assessing the immune response to SARS-CoV-2 infection. Methods Here, we combined two high-throughput immune profiling methods to create a quantitative picture of the T-cell response to SARS-CoV-2. First, at the individual level, we deeply characterized 3 acutely infected and 58 recovered COVID-19 subjects by experimentally mapping their CD8 T-cell response through antigen stimulation to 545 Human Leukocyte Antigen (HLA) class I presented viral peptides. Then, at the population level, we performed T-cell repertoire sequencing on 1,815 samples (from 1,521 COVID-19 subjects) as well as 3,500 controls to identify shared "public" T-cell receptors (TCRs) associated with SARS-CoV-2 infection from both CD8 and CD4 T cells. Results Collectively, our data reveal that CD8 T-cell responses are often driven by a few immunodominant, HLA-restricted epitopes. As expected, the T-cell response to SARS-CoV-2 peaks about one to two weeks after infection and is detectable for at least several months after recovery. As an application of these data, we trained a classifier to diagnose SARS-CoV-2 infection based solely on TCR sequencing from blood samples, and observed, at 99.8% specificity, high early sensitivity soon after diagnosis (Day 3-7 = 85.1% [95% CI = 79.9-89.7]; Day 8-14 = 94.8% [90.7-98.4]) as well as lasting sensitivity after recovery (Day 29+/convalescent = 95.4% [92.1-98.3]). Discussion The approaches described in this work provide detailed insights into the adaptive immune response to SARS-CoV-2 infection, and they have potential applications in clinical diagnostics, vaccine development, and monitoring.
Collapse
Affiliation(s)
| | | | - Mark Klinger
- Adaptive Biotechnologies, Seattle, WA, United States
| | - Damon H. May
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | | | | - Ian M. Kaplan
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | | - Ravi Pandya
- Microsoft Research, Redmond, WA, United States
| | - Xiaoyu Chen
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | | - Peter Ebert
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | | | | | | | | - Jennifer Lu
- Adaptive Biotechnologies, Seattle, WA, United States
| | - Allen Vong
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | - Paul Fields
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | | - Lance Baldo
- Adaptive Biotechnologies, Seattle, WA, United States
| | - Simona Semprini
- Unit of Microbiology - The Great Romagna Hub Laboratory, Pievesestina ITALY and DIMES, University of Bologna, Bologna, Italy
| | - Claudio Cerchione
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabio Nicolini
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Massimiliano Mazza
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Ottavia M. Delmonte
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Kerry Dobbs
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Rocio Laguna-Goya
- Department of Immunology, Hospital 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | | | - Santiago Barrio
- Hematology Department, Hospital 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | - Luisa Imberti
- Laboratorio CREA, Department of Infectious and Tropical Diseases, and Medical Officer, ASST Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | - Alessandra Sottini
- Laboratorio CREA, Department of Infectious and Tropical Diseases, and Medical Officer, ASST Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | - Eugenia Quiros-Roldan
- Laboratorio CREA, Department of Infectious and Tropical Diseases, and Medical Officer, ASST Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | - Camillo Rossi
- Laboratorio CREA, Department of Infectious and Tropical Diseases, and Medical Officer, ASST Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | - Andrea Biondi
- Department of Pediatrics and Centro Tettamanti-European Reference Network PaedCan, EuroBloodNet, MetabERN-University of Milano-Bicocca-Fondazione MBBM-Ospedale San Gerardo, Monza, Italy
| | - Laura Rachele Bettini
- Department of Pediatrics and Centro Tettamanti-European Reference Network PaedCan, EuroBloodNet, MetabERN-University of Milano-Bicocca-Fondazione MBBM-Ospedale San Gerardo, Monza, Italy
| | - Mariella D’Angio
- Department of Pediatrics and Centro Tettamanti-European Reference Network PaedCan, EuroBloodNet, MetabERN-University of Milano-Bicocca-Fondazione MBBM-Ospedale San Gerardo, Monza, Italy
| | - Paolo Bonfanti
- Department of Infectious Diseases, University of Milano-Bicocca-Ospedale San Gerardo, Monza, Italy
| | - Miranda F. Tompkins
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Camille Alba
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Clifton Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Vittorio Sambri
- Unit of Microbiology - The Great Romagna Hub Laboratory, Pievesestina ITALY and DIMES, University of Bologna, Bologna, Italy
| | - Giovanni Martinelli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Jason D. Goldman
- Swedish Medical Center, Seattle, WA, United States
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, United States
| | - James R. Heath
- Institute for Systems Biology, Seattle, WA, United States
| | - Helen C. Su
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Luigi D. Notarangelo
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Estela Paz-Artal
- Department of Immunology, Hospital 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | - Joaquin Martinez-Lopez
- Hematology Department, Hospital 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | - Bryan Howie
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | |
Collapse
|
6
|
Pedrazzoli S, Graziosi G, Salaroli R, Catelli E, Lupini C. Dynamic alterations in T-lymphocyte subsets assessed by flow cytometry in chickens following exposure to infectious bursal disease virus: A systematic review. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105280. [PMID: 39396690 DOI: 10.1016/j.dci.2024.105280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/22/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Infectious bursal disease virus (IBDV) is a significant pathogen in poultry, causing acute immunosuppressive disease in young chickens. While B-lymphocyte involvement in IBDV pathogenesis is known, the role of T-cells is incompletely understood. This systematic review presents the alterations in chicken T-lymphocyte subsets after IBDV exposure, assessed by flow cytometry analysis. Four databases were queried for identifying eligible studies focused on experimental infections measuring T-lymphocyte changes in the bursa of Fabricius, spleen, thymus, and peripheral blood mononuclear cells. Of 488 studies found, 25 met the pre-established criteria and were included in the qualitative synthesis of results. Most studies analysed T-lymphocyte responses during the acute phase of IBDV infection, primarily focusing on CD4+ and CD8+ T-cells. Other subsets, such as γδ T-cells and double-positive CD4+CD8+ T-cells, were less frequently investigated. An increase in T-lymphocytes was noted in the bursa of Fabricius, suggesting their active role in viral clearance. In the spleen, CD4+ T-cells commonly increased, while CD8+ responses varied among studies. Increased levels in T-cells were also noted during the chronic infection in the bursa of Fabricius, possibly due to persistent viral antigens. Overall, variations in flow cytometry methods and T-cell output reporting were noted among studies. Based on the data collected, further investigation into diverse T-cell subpopulations beyond CD4+ and CD8+ is needed, as well as the standardization of flow cytometry assays in chickens.
Collapse
Affiliation(s)
- Sara Pedrazzoli
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia BO, Italy.
| | - Giulia Graziosi
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia BO, Italy.
| | - Roberta Salaroli
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia BO, Italy.
| | - Elena Catelli
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia BO, Italy.
| | - Caterina Lupini
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia BO, Italy.
| |
Collapse
|
7
|
Shao Y, Gao Q, Wang L, Li D, Nixon AB, Chan C, Li QJ, Xie J. B-Lightning: using bait genes for marker gene hunting in single-cell data with complex heterogeneity. Brief Bioinform 2024; 26:bbaf033. [PMID: 39927857 PMCID: PMC11808808 DOI: 10.1093/bib/bbaf033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/12/2024] [Accepted: 02/03/2025] [Indexed: 02/11/2025] Open
Abstract
In single-cell studies, cells can be characterized with multiple sources of heterogeneity (SOH) such as cell type, developmental stage, cell cycle phase, activation state, and so on. In some studies, many nuisance SOH are of no interest, but may confound the identification of the SOH of interest, and thus affect the accurate annotate the corresponding cell subpopulations. In this paper, we develop B-Lightning, a novel and robust method designed to identify marker genes and cell subpopulations corresponding to an SOH (e.g. cell activation status), isolating it from other SOH (e.g. cell type, cell cycle phase). B-Lightning uses an iterative approach to enrich a small set of trustworthy marker genes to more reliable marker genes and boost the signals of the SOH of interest. Multiple numerical and experimental studies showed that B-Lightning outperforms existing methods in terms of sensitivity and robustness in identifying marker genes. Moreover, it increases the power to differentiate cell subpopulations of interest from other heterogeneous cohorts. B-Lightning successfully identified new senescence markers in ciliated cells from human idiopathic pulmonary fibrosis lung tissues, new T-cell memory and effector markers in the context of SARS-COV-2 infections, and their synchronized patterns that were previously neglected, new AD markers that can better differentiate AD severity, and new dendritic cell functioning markers with differential transcriptomics profiles across breast cancer subtypes. This paper highlights B-Lightning's potential as a powerful tool for single-cell data analysis, particularly in complex data sets where SOH of interest are entangled with numerous nuisance factors.
Collapse
Affiliation(s)
- Yiren Shao
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, United States
| | - Qi Gao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48104, United States
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708, United States
| | - Dongmei Li
- Department of Clinical and Translational Research, Unversity of Rochester Medical Center, Rochester, NY 14642, United States
| | - Andrew B Nixon
- Department of Medicine, Duke University, Durham, NC 27708, United States
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27708, United States
- Center for Human Systems Immunology, Duke University, Durham, NC 27708, United States
| | - Qi-Jing Li
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 138673, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research, 138648, Singapore
| | - Jichun Xie
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27708, United States
- Center for Human Systems Immunology, Duke University, Durham, NC 27708, United States
- Department of Mathematics, Duke University, Durham, NC 27708, United States
| |
Collapse
|
8
|
Shao Y, Gao Q, Wang L, Li D, Nixon AB, Chan C, Li QJ, Xie J. Marker gene fishing for single-cell data with complex heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.03.621735. [PMID: 39574750 PMCID: PMC11580937 DOI: 10.1101/2024.11.03.621735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
In single-cell studies, cells can be characterized with multiple sources of heterogeneity such as cell type, developmental stage, cell cycle phase, activation state, and so on. In some studies, many nuisance sources of heterogeneity (SOH) are of no interest, but may confound the identification of the SOH of interest, and thus affect the accurate annotate the corresponding cell subpopulations. In this paper, we develop B-Lightning, a novel and robust method designed to identify marker genes and cell subpopulations correponding to a SOH (e.g., cell activation status), isolating it from other sources of heterogeneity (e.g., cell type, cell cycle phase). B-Lightning uses an iterative approach to enrich a small set of trustworthy marker genes to more reliable marker genes and boost the signals of the SOH of interest. Multiple numerical and experimental studies showed that B-Lightning outperforms existing methods in terms of sensitivity and robustness in identifying marker genes. Moreover, it increases the power to differentiate cell subpopulations of interest from other heterogeneous cohorts. B-Lightning successfully identified new senescence markers in ciliated cells from human idiopathic pulmonary fibrosis (IPF) lung tissues, new T cell memory and effector markers in the context of SARS-COV-2 infections, and their synchronized patterns which were previously neglected. This paper highlights B-Lightning's potential as a powerful tool for single-cell data analysis, particularly in complex data sets where sources of heterogeneity of interest are entangled with numerous nuisance factors.
Collapse
|
9
|
Yazdan Panah M, Vaheb S, Mokary Y, Afshari-Safavi A, Shaygannejad A, Ebrahimi N, Shaygannejad V, Mirmosayyeb O. Comparing the consequences of COVID-19 vaccination between central nervous system (CNS) demyelinating diseases and other neurological disorders. Vaccine 2024; 42:126061. [PMID: 38886142 DOI: 10.1016/j.vaccine.2024.06.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/27/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Vaccination constitutes a crucial preventive measure against COVID-19 infection. Concerns have been raised regarding the efficacy of vaccines in multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) patients due to various immunomodulatory medications and potential adverse events that may impact neurological function. This study aimed to explore the implications of COVID-19 vaccination within MS and NMSOD patients and compare it with other neurological disorders (OND). METHOD In this cross-sectional study conducted in Isfahan, Iran, baseline data and information on COVID-19 infections and vaccinations were collected from MS, NMOSD, and OND patients between September 2021 and September 2022. The predominant neurological disorders identified among OND patients encompassed headache, epilepsy, and Parkinson's disease. Logistic regression analysis was employed to compare COVID-19 vaccination outcomes among different patient groups, presenting odds ratios (OR) with 95% confidence intervals (CI). RESULTS The study included 1,307 participants, with 738 having MS, 96 having NMOSD, 76 having clinically isolated syndrome (CIS), and 397 having OND. Significantly higher odds of post-vaccination COVID-19 infection were detected in MS (OR = 3.86, p < 0.001) NMOSD (OR = 2.77, p = 0.015) patients than OND patients. The prior history of COVID-19 infection and the type of vaccine administered did not demonstrate significant associations with the likelihood of post-vaccination COVID-19 infection in MS and NMOSD patients (p > 0.05 for all). There were no significant differences in the rates of adverse events in MS, NMOSD, and OND patients, except the second dose, where NMOSD patients had lower odds than OND patients (OR = 0.55, p = 0.019). CONCLUSION Although the safety profile of COVID-19 vaccination in MS and NMOSD was similar to that in OND, the rates of post-vaccination COVID-19 infection in MS and NMOSD seem higher than OND. These findings highlight the importance of regular serological monitoring and the potential advantages of supplementary vaccine doses in MS and NMOSD patients.
Collapse
Affiliation(s)
- Mohammad Yazdan Panah
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran; Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Vaheb
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yousef Mokary
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Afshari-Safavi
- Department of Biostatistics and Epidemiology, Faculty of Health, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Aysa Shaygannejad
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Narges Ebrahimi
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vahid Shaygannejad
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Omid Mirmosayyeb
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
10
|
Messiah SE, Abbas R, Bergqvist E, Kohl HW, Swartz MD, Talebi Y, Sabharwal R, Han H, Valerio-Shewmaker MA, DeSantis SM, Yaseen A, Gandhi HA, Amavisca XF, Ross JA, Padilla LN, Gonzalez MO, Wu L, Silberman MA, Lakey D, Shuford JA, Pont SJ, Boerwinkle E. Factors associated with elevated SARS-CoV-2 immune response in children and adolescents. Front Pediatr 2024; 12:1393321. [PMID: 39228441 PMCID: PMC11369978 DOI: 10.3389/fped.2024.1393321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/22/2024] [Indexed: 09/05/2024] Open
Abstract
Background Understanding the distinct immunologic responses to SARS-CoV-2 infection among pediatric populations is pivotal in navigating the COVID-19 pandemic and informing future public health strategies. This study aimed to identify factors associated with heightened antibody responses in children and adolescents to identify potential unique immune dynamics in this population. Methods Data collected between July and December 2023 from the Texas Coronavirus Antibody REsponse Survey (Texas CARES), a statewide prospective population-based antibody survey among 1-to-19-year-old participants, were analyzed. Each participant had the following data available for analysis: (1) Roche Elecsys® Anti-SARS-CoV-2 Immunoassay for Nucleocapsid protein antibodies (Roche N-test), (2) qualitative and semi-quantitative detection of antibodies to the SARS CoV-2 spike protein receptor binding domain (Roche S-test), and (3) self-reported antigen/PCR COVID-19 test results, vaccination, and health status. Statistical analysis identified associations between participant characteristics and spike antibody quartile group. Results The analytical sample consisted of 411 participants (mean age 12.2 years, 50.6% female). Spike antibody values ranged from a low of 6.3 U/ml in the lowest quartile to a maximum of 203,132.0 U/ml in the highest quartile in the aggregate sample. Older age at test date (OR = 1.22, 95% CI: 1.12, 1.35, p < .001) and vaccination status (primary series/partially vaccinated, one or multiple boosters) showed significantly higher odds of being in the highest spike antibody quartile compared to younger age and unvaccinated status. Conversely, fewer days since the last immunity challenge showed decreased odds (OR = 0.98, 95% CI: 0.96, 0.99, p = 0.002) of being in the highest spike antibody quartile vs. more days since last immunity challenge. Additionally, one out of every three COVID-19 infections were asymptomatic. Conclusions Older age, duration since the last immunity challenge (vaccine or infection), and vaccination status were associated with heightened spike antibody responses, highlighting the nuanced immune dynamics in the pediatric population. A significant proportion of children/adolescents continue to have asymptomatic infection, which has important public health implications.
Collapse
Affiliation(s)
- Sarah E. Messiah
- Department of Epidemiology, UTHealth Houston School of Public Health, Dallas, TX, United States
- Center for Pediatric Population Health, UTHealth Houston School of Public Health, Dallas, TX, United States
- Department of Pediatrics, McGovern Medical School at UTHealth Houston, Houston, TX, United States
| | - Rhiana Abbas
- Department of Biostatistics and Data Science, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Emma Bergqvist
- Department of Epidemiology, UTHealth Houston School of Public Health, Dallas, TX, United States
- Center for Pediatric Population Health, UTHealth Houston School of Public Health, Dallas, TX, United States
| | - Harold W. Kohl
- Department of Epidemiology, UTHealth Houston School of Public Health, Austin, TX, United States
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, United States
| | - Michael D. Swartz
- Department of Biostatistics and Data Science, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Yashar Talebi
- Department of Biostatistics and Data Science, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Rachit Sabharwal
- Department of Biostatistics and Data Science, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Haoting Han
- Department of Biostatistics and Data Science, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Melissa A. Valerio-Shewmaker
- Department of Health Promotion and Behavioral Sciences, UTHealth Houston School of Public Health, Brownsville, TX, United States
| | - Stacia M. DeSantis
- Department of Biostatistics and Data Science, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Ashraf Yaseen
- Department of Biostatistics and Data Science, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Henal A. Gandhi
- Department of Epidemiology, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Ximena Flandes Amavisca
- Department of Epidemiology, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Jessica A. Ross
- Department of Epidemiology, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Lindsay N. Padilla
- Department of Epidemiology, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Michael O. Gonzalez
- Department of Biostatistics and Data Science, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Leqing Wu
- Department of Biostatistics and Data Science, UTHealth Houston School of Public Health, Houston, TX, United States
| | | | - David Lakey
- The University of Texas System, Austin, TX, United States
- The University of Texas at Tyler Health Science Center, Tyler, TX, United States
| | | | - Stephen J. Pont
- Texas Department of State Health Services, Austin, TX, United States
| | - Eric Boerwinkle
- Department of Epidemiology, UTHealth Houston School of Public Health, Houston, TX, United States
| |
Collapse
|
11
|
Carretero D, Giménez E, Albert E, Colomer E, Montomoli M, Hernani R, Piñana JL, Górriz JL, Solano C, Navarro D. Performance of the Roche Elecsys® IGRA SARS-CoV-2 test for the detection and quantification of virus-reactive T cells in COVID-19-vaccinated immunosuppressed patients and healthy subjects. Eur J Clin Microbiol Infect Dis 2024; 43:1427-1436. [PMID: 38777919 PMCID: PMC11271317 DOI: 10.1007/s10096-024-04852-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE Comparing the performance of commercially available SARS-CoV-2 T-cell immunoassay responses may provide useful information for future observational or intervention studies as well as to their potential customers. METHOD Whole blood was collected from a total of 183 subjects fully vaccinated against COVID-19: 55 healthy controls (Group 1), 50 hematological patients (Group 2), 50 chronic kidney disease patients (Group 3), and 28 elderly nursing home residents (Group 4). Samples were tested with the Roche Elecsys® IGRA (Interferon-gamma release assay) SARS-CoV-2 test (Roche Diagnostics, Rotkreuz, Switzerland), the Euroimmun SARS-CoV-2 test (Euroimmun, Lubeck, Germany), the SARS-CoV-2 T Cell Analysis Kit (Miltenyi Biotec, Bergisch Gladbach, Germany), and a flow-cytometry for intracellular cytokine (IFN-γ) staining-based immunoassay (FC-ICS). RESULTS Overall, the Roche Elecsys® assay returned the highest number of positive results (151/179; 84.3%), followed by the Euroimmun test (127/183; 69%), and the FC-ICS (135/179; 75%). The Kappa coefficient of agreement was best between IGRAs (0.64). Most discordant results across assays involved patients from Group 2. Overall, IFN-γ concentrations measured by both IGRAs correlated strongly (rho = 0.78; 95% CI 0.71-0.84; P < 0.001) irrespective of the study group. The frequencies of SARS-CoV-2-reactive IFN-γ T cells and IFN-γ concentrations measured by the IGRAs correlated moderately for CD4+ T cells, however, weakly for CD8+ T cells. SARS-CoV-2-experienced participants displayed stronger responses than SARS-CoV-2-naïve when IGRAs, rather than FC-ICS, were used. CONCLUSION The SARS-CoV-2 immunoassays evaluated in the present study did not return interchangeable qualitative or quantitative results either in seemingly healthy individuals or in immunosuppressed patients.
Collapse
Affiliation(s)
- Diego Carretero
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Estela Giménez
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Eliseo Albert
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Ester Colomer
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Marco Montomoli
- Nephrology Service, Hospital Clínico Universitario de Valencia, INCLIVA Health Research Institute, Valencia, Spain
| | - Rafael Hernani
- Hematology Service, Hospital Clínico Universitario, INCLIVA Health Research Institute, Valencia, Spain
| | - José Luis Piñana
- Hematology Service, Hospital Clínico Universitario, INCLIVA Health Research Institute, Valencia, Spain
| | - José Luis Górriz
- Nephrology Service, Hospital Clínico Universitario de Valencia, INCLIVA Health Research Institute, Valencia, Spain
- Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain
| | - Carlos Solano
- Hematology Service, Hospital Clínico Universitario, INCLIVA Health Research Institute, Valencia, Spain
- Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain
| | - David Navarro
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain.
- Department of Microbiology, School of Medicine, University of Valencia, Valencia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
- Microbiology Service, Hospital Clínico Universitario, Instituto de Investigación INCLIVA, Av. Blasco Ibáñez 17, 46010, Valencia, Spain.
| |
Collapse
|
12
|
Felisberto M, Walter LO, Cardoso CC, Santos-Pirath ÍM, Costa HZ, Gartner R, Werle I, Mohr ETB, Salvan da Rosa J, Lubschinski TL, Kretzer IF, Masukawa II, de Almeida Vanny P, Luiz MC, Rabello de Moraes AC, Santos-Silva MC, Dalmarco EM. Lymphocyte B Subtypes in Peripheral Blood: A Prognostic Biomarker for COVID-19 Patients. J Appl Lab Med 2024; 9:456-467. [PMID: 38321537 DOI: 10.1093/jalm/jfad123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND In view of the scientific gap in knowledge of the involvement of the B-cell compartment and clinical prognostic in SARS-CoV-2 infection, this work aims to evaluate the B-cell subsets and the presence of specific IgM and IgG, as well as neutralizing antibodies against SARS-CoV-2, in unvaccinated patients diagnosed with COVID-19. METHODS This study included 133 patients with COVID-19. Cellular components were assessed by flow cytometry, and immunoglobulin levels and reactivity were measured by indirect enzyme-linked immunosorbent assay. RESULTS Our results showed no changes in less differentiated B cells. However, non-switched memory B cells (NS-MBCs) and class-switched memory B cells (CS-MBCs) were reduced in the patients with moderate disease. Also, plasmablasts and double-negative (DN) or "atypical" memory B cells were increased in groups of patients with moderate to critical conditions. In addition, the production of IgM, IgG, and neutralizing antibodies against SARS-CoV-2 demonstrated a positive correlation between the positivity of antibodies against SARS-CoV-2 and disease severity. Besides being related to the development of a more severe course of the disease, the increase in DN B-cell count also contributed to a poorer disease outcome in patients with a higher percentage of these cells. On the other hand, we observed an increase in the absolute number of CS-MBCs in patients with greater chances of survival. CONCLUSIONS This study demonstrates that the B-cell compartment may contribute to the development of clinical symptoms of COVID-19, with changes in B-cell subset counts linked to disease course and patient prognosis.
Collapse
Affiliation(s)
- Mariano Felisberto
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Laura Otto Walter
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Chandra Chiappin Cardoso
- Clinical Analysis Department, Flow Cytometry Service, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Íris Mattos Santos-Pirath
- Clinical Analysis Department, Flow Cytometry Service, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Heloisa Zorzi Costa
- Clinical Analysis Department, Flow Cytometry Service, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Rafaela Gartner
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Isabel Werle
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Eduarda Talita Bramorski Mohr
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Julia Salvan da Rosa
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Tainá Larissa Lubschinski
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Iara Fabricia Kretzer
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Ivete Ioshiko Masukawa
- Infectious Disease Service, University Hospital-Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Infectious Disease Service, State Health Department, Hospital Nereu Ramos, Florianópolis, SC, Brazil
| | - Patrícia de Almeida Vanny
- Infectious Disease Service, University Hospital-Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Magali Chaves Luiz
- Infectious Disease Service, State Health Department, Hospital Nereu Ramos, Florianópolis, SC, Brazil
| | - Ana Carolina Rabello de Moraes
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Maria Claudia Santos-Silva
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Flow Cytometry Service, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Eduardo Monguilhott Dalmarco
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| |
Collapse
|
13
|
Giorda CB, Picariello R, Tartaglino B, Nada E, Romeo F, Costa G, Gnavi R. Hospitalisation for herpes zoster in people with and without diabetes: A 10-year-observational study. Diabetes Res Clin Pract 2024; 210:111603. [PMID: 38460790 DOI: 10.1016/j.diabres.2024.111603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/11/2024]
Abstract
AIMS This study explores the association between Herpes Zoster (HZ) hospitalizations and diabetes in Piedmont, Italy from 2010 to 2019. Focusing on the burden of HZ hospitalizations in diabetic and non-diabetic groups, it aims to identify risk factors in diabetics to enhance prevention strategies. METHODS In a two-phase study, we first compared age-standardized HZ hospitalization rates between diabetic and non-diabetic individuals from 2010 to 2019. We then examined hospitalization risk factors for HZ within a diabetic patient cohort managed by regional diabetes clinics. RESULTS Of 3,423 HZ hospitalizations in 2010-2019, 17.9 % (613 cases) were diabetic patients, who exhibited higher hospitalization rates (15.9 to 6.0 per 100,000) compared to non-diabetese individuals. Among diabetics subjects risk factors for HZ hospitalization included age over 65, obesity (BMI > 30), and poor glycemic control (HbA1c > 8.0 %). These patients had a 40 % increased rehospitalization risk and a 25 % higher risk of severe complications, such as stroke and myocardial infarction, post-HZ. CONCLUSIONS Diabetes markedly increases HZ hospitalization rates, rehospitalization, and complication risks. These findings underscore the need for preventive strategies, especially improved glycemic control among high-risk diabetic patients, to inform public health policies and clinical practices aimed at mitigating HZ's impact on this population.
Collapse
Affiliation(s)
- Carlo B Giorda
- Metabolism and Diabetes Unit, ASL TO5, Regione Piemonte, Chieri, Italy.
| | | | | | - Elisa Nada
- Metabolism and Diabetes Unit, ASL TO5, Regione Piemonte, Chieri, Italy
| | - Francesco Romeo
- Metabolism and Diabetes Unit, ASL TO5, Regione Piemonte, Chieri, Italy
| | - Giuseppe Costa
- Epidemiology Unit, ASL TO3, Regione Piemonte, Grugliasco, Italy; Department of Public Health, University of Torino, Torino, Italy
| | - Roberto Gnavi
- Epidemiology Unit, ASL TO3, Regione Piemonte, Grugliasco, Italy
| |
Collapse
|
14
|
Ng’uni TL, Musale V, Nkosi T, Mandolo J, Mvula M, Michelo C, Karim F, Moosa MYS, Khan K, Jambo KC, Hanekom W, Sigal A, Kilembe W, Ndhlovu ZM. Low pre-existing endemic human coronavirus (HCoV-NL63)-specific T cell frequencies are associated with impaired SARS-CoV-2-specific T cell responses in people living with HIV. Front Immunol 2024; 14:1291048. [PMID: 38343437 PMCID: PMC10853422 DOI: 10.3389/fimmu.2023.1291048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/18/2023] [Indexed: 02/15/2024] Open
Abstract
Background Understanding how HIV affects SARS-CoV-2 immunity is crucial for managing COVID-19 in sub-Saharan populations due to frequent coinfections. Our previous research showed that unsuppressed HIV is associated with weaker immune responses to SARS-CoV-2, but the underlying mechanisms are unclear. We investigated how pre-existing T cell immunity against an endemic human coronavirus HCoV-NL63 impacts SARS-CoV-2 T cell responses in people living with HIV (PLWH) compared to uninfected individuals, and how HIV-related T cell dysfunction influences responses to SARS-CoV-2 variants. Methods We used flow cytometry to measure T cell responses following PBMC stimulation with peptide pools representing beta, delta, wild-type, and HCoV-NL63 spike proteins. Luminex bead assay was used to measure circulating plasma chemokine and cytokine levels. ELISA and MSD V-PLEX COVID-19 Serology and ACE2 Neutralization assays were used to measure humoral responses. Results Regardless of HIV status, we found a strong positive correlation between responses to HCoV-NL63 and SARS-CoV-2. However, PLWH exhibited weaker CD4+ T cell responses to both HCoV-NL63 and SARS-CoV-2 than HIV-uninfected individuals. PLWH also had higher proportions of functionally exhausted (PD-1high) CD4+ T cells producing fewer proinflammatory cytokines (IFNγ and TNFα) and had elevated plasma IL-2 and IL-12(p70) levels compared to HIV-uninfected individuals. HIV status didn't significantly affect IgG antibody levels against SARS-CoV-2 antigens or ACE2 binding inhibition activity. Conclusion Our results indicate that the decrease in SARS-CoV-2 specific T cell responses in PLWH may be attributable to reduced frequencies of pre-existing cross-reactive responses. However, HIV infection minimally affected the quality and magnitude of humoral responses, and this could explain why the risk of severe COVID-19 in PLWH is highly heterogeneous.
Collapse
Affiliation(s)
- Tiza L. Ng’uni
- Africa Health Research Institute (AHRI), Nelson R. Mandela School of Medicine, Durban, South Africa
| | - Vernon Musale
- Emory-University of Georgia, Center of Excellence of Influenza Research and Surveillance (CEIRS), Lusaka, Zambia
- Center for Family Health Research in Zambia (CFHRZ), formerly Zambia Emory HIV Research Project (ZEHRP), Lusaka, Zambia
| | - Thandeka Nkosi
- Africa Health Research Institute (AHRI), Nelson R. Mandela School of Medicine, Durban, South Africa
| | - Jonathan Mandolo
- Infection and Immunity Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Memory Mvula
- Infection and Immunity Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Clive Michelo
- Emory-University of Georgia, Center of Excellence of Influenza Research and Surveillance (CEIRS), Lusaka, Zambia
- Center for Family Health Research in Zambia (CFHRZ), formerly Zambia Emory HIV Research Project (ZEHRP), Lusaka, Zambia
| | - Farina Karim
- Africa Health Research Institute (AHRI), Nelson R. Mandela School of Medicine, Durban, South Africa
| | - Mohomed Yunus S. Moosa
- Human Immunodeficiency Virus (HIV) Pathogenesis Program, School of Laboratory Medicine and Medical Sciences, University of KwaZulu Natal, Durban, South Africa
| | - Khadija Khan
- Africa Health Research Institute (AHRI), Nelson R. Mandela School of Medicine, Durban, South Africa
| | - Kondwani Charles Jambo
- Infection and Immunity Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Willem Hanekom
- Africa Health Research Institute (AHRI), Nelson R. Mandela School of Medicine, Durban, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Alex Sigal
- Africa Health Research Institute (AHRI), Nelson R. Mandela School of Medicine, Durban, South Africa
| | - William Kilembe
- Emory-University of Georgia, Center of Excellence of Influenza Research and Surveillance (CEIRS), Lusaka, Zambia
- Center for Family Health Research in Zambia (CFHRZ), formerly Zambia Emory HIV Research Project (ZEHRP), Lusaka, Zambia
| | - Zaza M. Ndhlovu
- Africa Health Research Institute (AHRI), Nelson R. Mandela School of Medicine, Durban, South Africa
- Human Immunodeficiency Virus (HIV) Pathogenesis Program, School of Laboratory Medicine and Medical Sciences, University of KwaZulu Natal, Durban, South Africa
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard University, Cambridge, MA, United States
| |
Collapse
|
15
|
Masiá M, de la Rica A, Fernández-González M, García JA, Padilla S, García-Abellán J, Botella Á, Mascarell P, Gutiérrez F. Integrating SARS-CoV-2-specific interferon-γ release assay testing in the evaluation of patients hospitalized with COVID-19. Microbiol Spectr 2023; 11:e0241923. [PMID: 37855635 PMCID: PMC10715100 DOI: 10.1128/spectrum.02419-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/06/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE The cellular immune response is essential in the protection against severe disease in patients with established SARS-CoV-2 infection. The novelty of this study lies in the evaluation of the overall performance of a standardized assay to measure cellular immune response, the SARS-CoV-2-specific interferon-γ release assay (IGRA), in hospitalized patients with severe COVID-19. The SARS-CoV-2 IGRA was shown to accurately classify patients based on disease severity and prognosis, and the study revealed that test performance was not affected by the SARS-CoV-2 variant or control tube results. We identified an assay cut-off point with a high negative predictive value against mortality. The SARS-CoV-2 IGRA in patients hospitalized for COVID-19 may be a useful tool to assess cellular immunity and adopt targeted therapeutic and preventive measures.
Collapse
Affiliation(s)
- Mar Masiá
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Alba de la Rica
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- Microbiology Service, Hospital General Universitario de Elche, Alicant, Spain
| | - Marta Fernández-González
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - José Alberto García
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Padilla
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Javier García-Abellán
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Ángela Botella
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
| | - Paula Mascarell
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
| | - Félix Gutiérrez
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| |
Collapse
|
16
|
Plano F, Shekarkar Azgomi M, Corsale AM, Spoto C, Caccamo N, Meraviglia S, Dieli F, D’Angelo P, Trizzino A, Siragusa S. Humoral and Cell-Mediated Responses to SARS-CoV-2 Vaccination in a Cohort of Immunodeficient Patients. Hematol Rep 2023; 15:707-716. [PMID: 38132279 PMCID: PMC10742827 DOI: 10.3390/hematolrep15040071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
This study delves into the intricate landscape of SARS-CoV-2 vaccine response in immunodeficient patients, focusing on the dynamics of both humoral and cell-mediated immunity. The cohort includes patients with common variable immunodeficiency (CVI), agammaglobulinemia (XLA), and combined immunodeficiency (CI). The findings reveal varying degrees of antibody production, with XLA patients exhibiting no measurable response but displaying a robust T-cell-mediated response. The study emphasizes the importance of considering both arms of the immune system in assessing vaccine immunogenicity, particularly in the context of immunodeficiency. The results challenge conventional measures of vaccine efficacy only based on antibody titers, highlighting the need for a more comprehensive understanding of the immune response in this vulnerable population. This research contributes valuable insights to guide clinical decisions regarding vaccination strategies, booster doses, and overall protection in immunodeficient individuals.
Collapse
Affiliation(s)
- Federica Plano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.M.C.); (C.S.); (S.S.)
| | - Mojtaba Shekarkar Azgomi
- Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, 90127 Palermo, Italy; (M.S.A.); (N.C.); (S.M.); (F.D.)
| | - Anna Maria Corsale
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.M.C.); (C.S.); (S.S.)
| | - Corinne Spoto
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.M.C.); (C.S.); (S.S.)
| | - Nadia Caccamo
- Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, 90127 Palermo, Italy; (M.S.A.); (N.C.); (S.M.); (F.D.)
| | - Serena Meraviglia
- Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, 90127 Palermo, Italy; (M.S.A.); (N.C.); (S.M.); (F.D.)
| | - Francesco Dieli
- Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, 90127 Palermo, Italy; (M.S.A.); (N.C.); (S.M.); (F.D.)
| | - Paolo D’Angelo
- Department of Pediatric Hemato-Oncology, ARNAS Ospedali Civico, G. Di Cristina, 90127 Palermo, Italy; (P.D.); (A.T.)
| | - Antonino Trizzino
- Department of Pediatric Hemato-Oncology, ARNAS Ospedali Civico, G. Di Cristina, 90127 Palermo, Italy; (P.D.); (A.T.)
| | - Sergio Siragusa
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.M.C.); (C.S.); (S.S.)
| |
Collapse
|
17
|
Miller C, Taylor-Salmon E, Emuren L, Landry M, Gershon A, Miller G. Progressive shingles in a toddler due to reactivation of Varicella Zoster vaccine virus four days after infection with SARS-CoV-2; a case report. BMC Infect Dis 2023; 23:854. [PMID: 38057696 PMCID: PMC10698951 DOI: 10.1186/s12879-023-08809-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 11/09/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Herpes zoster (HZ) is the clinical syndrome associated with reactivation of latent varicella-zoster virus (VZV). Several factors have been implicated to promote VZV reactivation; these include immunosuppression, older age, mechanical trauma, physiologic stress, lymphopenia, and more recently, infection with severe acute respiratory syndrome coronavirus-2 (SARS- CoV-2). Recent reports suggest an increase in the number of HZ cases in the general population during the global COVID-19 pandemic. However, it is unknown what proportion of HZ during the pandemic is due to reactivation of wild-type or vaccine-strain VZV. CASE Here we report the first known case of HZ concomitant with SARS-CoV2 infection in a 20-month-old female who was treated with a single dose of dexamethasone, due to reactivation of the vaccine-type strain of VZV after presenting with a worsening vesicular rash. CONCLUSION In this case, we were able to show vaccine-strain VZV reactivation in the context of a mild acute symptomatic COVID-19 infection in a toddler. Being able to recognize HZ quickly and effectively in a pediatric patient can help stave off the significant morbidity and mortality associated with disease process.
Collapse
Affiliation(s)
- Christine Miller
- Department of Pediatrics, Section of Infectious Diseases and Global Health, Yale University School of Medicine, 464 Congress Ave, New Haven, CT, 06519, USA
| | - Emma Taylor-Salmon
- Department of Pediatrics, Section of Infectious Diseases and Global Health, Yale University School of Medicine, 464 Congress Ave, New Haven, CT, 06519, USA
| | - Leonard Emuren
- Department of Pediatrics, Section of Infectious Diseases and Global Health, Yale University School of Medicine, 464 Congress Ave, New Haven, CT, 06519, USA
| | - Marie Landry
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Anne Gershon
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - George Miller
- Department of Pediatrics, Section of Infectious Diseases and Global Health, Yale University School of Medicine, 464 Congress Ave, New Haven, CT, 06519, USA.
| |
Collapse
|
18
|
Ko GY, Lee J, Bae H, Ryu JH, Park HS, Kang H, Jung J, Choi AR, Lee R, Lee DG, Oh EJ. Longitudinal Analysis of SARS-CoV-2-Specific Cellular and Humoral Immune Responses and Breakthrough Infection following BNT162b2/BNT162b2/BNT162b2 and ChAdOx1/ChAdOx1/BNT162b2 Vaccination: A Prospective Cohort in Naive Healthcare Workers. Vaccines (Basel) 2023; 11:1613. [PMID: 37897015 PMCID: PMC10610978 DOI: 10.3390/vaccines11101613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/02/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Assessing immune responses post-SARS-CoV-2 vaccination is crucial for optimizing vaccine strategies. This prospective study aims to evaluate immune responses and breakthrough infection in 235 infection-naïve healthcare workers up to 13-15 months after initial vaccination in two vaccine groups (108 BNT/BNT/BNT and 127 ChAd/ChAd/BNT). Immune responses were assessed using the interferon-gamma enzyme-linked immunospot (ELISPOT) assay, total immunoglobulin, and neutralizing activity through surrogate virus neutralization test at nine different time points. Both groups exhibited peak responses one to two months after the second or third dose, followed by gradual declines over six months. Notably, the ChAd group exhibited a gradual increase in ELISPOT results, but their antibody levels declined more rapidly after reaching peak response compared to the BNT group. Six months after the third dose, both groups had substantial cellular responses, with superior humoral responses in the BNT group (p < 0.05). As many as 55 breakthrough infection participants displayed higher neutralization activities against Omicron variants, but similar cellular responses compared to 127 infection-naïve individuals, suggesting cross-immunity. Distinct neutralization classifications (<30%, >80% inhibition) correlated with different ELISPOT results. Our study reveals diverse immune response patterns based on vaccine strategies and breakthrough infections, emphasizing the importance of understanding these dynamics for optimized vaccination decisions.
Collapse
Affiliation(s)
- Geon Young Ko
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea; (G.Y.K.); (J.L.); (H.B.)
| | - Jihyun Lee
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea; (G.Y.K.); (J.L.); (H.B.)
| | - Hyunjoo Bae
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea; (G.Y.K.); (J.L.); (H.B.)
| | - Ji Hyeong Ryu
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.H.R.); (H.-S.P.); (H.K.); (J.J.); (A.-R.C.)
| | - Hye-Sun Park
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.H.R.); (H.-S.P.); (H.K.); (J.J.); (A.-R.C.)
| | - Hyunhye Kang
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.H.R.); (H.-S.P.); (H.K.); (J.J.); (A.-R.C.)
- Resesarch and Development Institute for In Vitro Diagnostic Medical Devices, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin Jung
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.H.R.); (H.-S.P.); (H.K.); (J.J.); (A.-R.C.)
- Resesarch and Development Institute for In Vitro Diagnostic Medical Devices, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Ae-Ran Choi
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.H.R.); (H.-S.P.); (H.K.); (J.J.); (A.-R.C.)
| | - Raeseok Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (R.L.); (D.-G.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Dong-Gun Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (R.L.); (D.-G.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Eun-Jee Oh
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.H.R.); (H.-S.P.); (H.K.); (J.J.); (A.-R.C.)
- Resesarch and Development Institute for In Vitro Diagnostic Medical Devices, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
19
|
Ravindran R, O’Connor E, Gupta A, Luciw PA, Khan AI, Dorreh N, Chiang K, Ikram A, Reddy S. Lipid Mediators and Cytokines/Chemokines Display Differential Profiles in Severe versus Mild/Moderate COVID-19 Patients. Int J Mol Sci 2023; 24:13054. [PMID: 37685858 PMCID: PMC10488250 DOI: 10.3390/ijms241713054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Host immune responses play a key role in COVID-19 pathogenesis. The underlying phenomena are orchestrated by signaling molecules such as cytokines/chemokines and lipid mediators. These immune molecules, including anti-SARS-CoV-2 antibodies, interact with immune cells and regulate host responses, contributing to inflammation that drives the disease. We investigated 48 plasma cytokines/chemokines, 21 lipid mediators, and anti-S protein (RBD) antibodies in COVID-19 patients (n = 56) and non-COVID-19 respiratory disease controls (n = 49), to identify immune-biomarker profiles. Cytokines/chemokines (IL-6, CXCL-10 (IP-10), HGF, MIG, MCP-1, and G-CSF) and lipid mediators (TxB2, 11-HETE, 9-HODE, 13-HODE, 5-HETE, 12-HETE, 15-HETE, 14S-HDHA, 17S-HDHA, and 5-oxo ETE) were significantly elevated in COVID-19 patients compared to controls. In patients exhibiting severe disease, pro-inflammatory cytokines/chemokines (IL-6, CXCL-10, and HGF) and anti-SARS-CoV-2 antibodies were significantly elevated. In contrast, lipid mediators involved in the reduction/resolution of inflammation, in particular, 5-HETE, 11-HETE, and 5-oxoETE, were significantly elevated in mild/moderate disease. Taken together, these immune-biomarker profiles provide insight into immune responses related to COVID-19 pathogenesis. Importantly, our findings suggest that elevation in plasma concentrations of IL-6, CXCL-10, HGF, and anti-SARS-CoV-2 antibodies can predict severe disease, whereas elevation in lipid mediators peaks early (compared to cytokines) and includes induction of mechanisms leading to reduction of inflammation, associated complications, and maintenance of homeostasis.
Collapse
Affiliation(s)
- Resmi Ravindran
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA 95817, USA;
| | - Ellen O’Connor
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (E.O.); (N.D.); (K.C.)
| | - Ajay Gupta
- Division of Nephrology, Hypertension and Kidney Transplantation, Department of Medicine, University of California Irvine (UCI) School of Medicine, Irvine, CA 92868, USA;
| | - Paul A. Luciw
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA 95817, USA;
| | - Aleena I. Khan
- Department of Population and Public Health, Keek School of Medicine, University of Southern California, Los Angeles, CA 90089, USA;
| | - Nasrin Dorreh
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (E.O.); (N.D.); (K.C.)
| | - Kate Chiang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (E.O.); (N.D.); (K.C.)
| | - Aamer Ikram
- National Institutes of Health, Islamabad 45500, Pakistan;
| | - Srinivasa Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (E.O.); (N.D.); (K.C.)
| |
Collapse
|
20
|
Pitiriga VC, Papamentzelopoulou M, Konstantinakou KE, Theodoridou K, Vasileiou IV, Tsakris A. SARS-CoV-2 T Cell Immunity Responses following Natural Infection and Vaccination. Vaccines (Basel) 2023; 11:1186. [PMID: 37515000 PMCID: PMC10384199 DOI: 10.3390/vaccines11071186] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
(1) Background: SARS-CoV-2 T cell immunity is rapidly activated following SARS-CoV-2 infection and vaccination and is crucial for controlling infection progression and severity. The aim of the present study was to compare the levels of T cell responses to SARS-CoV-2 between cohorts of subjects with hybrid immunity (convalescent and vaccinated), vaccinated naïve (non-exposed) and convalescent unvaccinated subjects. (2) Methods: We performed a retrospective descriptive analysis of data collected from the medical records of adult individuals who were consecutively examined at a large, private Medical Center of Attica from September 2021 to September 2022 in order to be examined on their own initiative for SARS-CoV-2 T cell immunity response. They were divided into three groups: Group A: SARS-CoV-2 convalescent and vaccinated subjects; Group B: SARS-CoV-2 naïve vaccinated subjects; Group C: SARS-CoV-2 convalescent unvaccinated subjects. The SARS-CoV-2 T cell response was estimated against spike (S) and nucleocapsid (N) structural proteins by performing the methodology T-SPOT.COVID test. (3) Results: A total of 530 subjects were retrospectively included in the study, 252 females (47.5%) and 278 (52.5%) males ranging from 13 to 92 years old (mean 55.68 ± 17.0 years). Among them, 66 (12.5%) were included in Group A, 284 (53.6%) in Group B and 180 (34.0%) in Group C. Among the three groups, a reaction against S antigen was reported in 58/66 (87.8%) of Group A, 175/284 (61.6%) of Group B and 146/180 (81.1%) of Group C (chi-square, p < 0.001). Reaction against N antigen was present in 49/66 (74.2%) of Group A and in 140/180 (77.7%) of Group C (chi-square, p = 0.841). The median SFC count for S antigen was 24 (range from 0-218) in Group A, 12 (range from 0-275) in Group B and 18 (range from 0-160) in Group C (Kruskal-Wallis test, p < 0.001; pairwise comparisons: groups A-B, p < 0.001; groups A-C, p = 0.147; groups B-C, p < 0.001). The median SFCs count for N antigen was 13 (range 0-82) for Group A and 18 (range 0-168) for Group C (Kruskal-Wallis test, p = 0.27 for A-C groups). (4) Conclusions: Our findings suggest that natural cellular immunity, either alone or combined with vaccination, confers stronger and more durable protection compared to vaccine-induced cellular immunity.
Collapse
Affiliation(s)
- Vassiliki C Pitiriga
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece
| | - Myrto Papamentzelopoulou
- Molecular Biology Unit, 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece
| | | | - Kalliopi Theodoridou
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece
| | - Irene V Vasileiou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece
| |
Collapse
|
21
|
Filippini F, Giacomelli M, Bazzani C, Fredi M, Semeraro P, Tomasi C, Franceschini F, Caruso A, Cavazzana I, Giagulli C. Efficacy of COVID-19 mRNA vaccination in patients with autoimmune disorders: humoral and cellular immune response. BMC Med 2023; 21:210. [PMID: 37316832 DOI: 10.1186/s12916-023-02868-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 04/17/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND The impact of immunosuppressive therapies on the efficacy of vaccines to SARS-CoV-2 is not completely clarified. We analyzed humoral and T cell-mediated response after COVID-19 mRNA vaccine in immunosuppressed patients and patients with common variable immunodeficiency disease (CVID). PATIENTS We enrolled 38 patients and 11 healthy sex- and age-matched controls (HC). Four patients were affected by CVID and 34 by chronic rheumatic diseases (RDs). All patients with RDs were treated by corticosteroid therapy and/or immunosuppressive treatment and/or biological drugs: 14 patients were treated with abatacept, 10 with rituximab, and 10 with tocilizumab. METHODS Total antibody titer to SARS-CoV-2 spike protein was assessed by electrochemiluminescence immunoassay, CD4 and CD4-CD8 T cell-mediated immune response was analyzed by interferon-γ (IFN-γ) release assay, the production of IFN-γ-inducible (CXCL9 and CXCL10) and innate-immunity chemokines (MCP-1, CXCL8, and CCL5) by cytometric bead array after stimulation with different spike peptides. The expression of CD40L, CD137, IL-2, IFN-γ, and IL-17 on CD4 and CD8 T cells, evaluating their activation status, after SARS-CoV-2 spike peptides stimulation, was analyzed by intracellular flow cytometry staining. Cluster analysis identified cluster 1, namely the "high immunosuppression" cluster, and cluster 2, namely the "low immunosuppression" cluster. RESULTS After the second dose of vaccine, only abatacept-treated patients, compared to HC, showed a reduced anti-spike antibody response (mean: 432 IU/ml ± 562 vs mean: 1479 IU/ml ± 1051: p = 0.0034), and an impaired T cell response, compared with HC. In particular, we found a significantly reduced release of IFN-γ from CD4 and CD4-CD8 stimulated T cells, compared with HC (p = 0.0016 and p = 0.0078, respectively), reduced production of CXCL10 and CXCL9 from stimulated CD4 (p = 0.0048 and p = 0.001) and CD4-CD8 T cells (p = 0.0079 and p = 0.0006). Multivariable General Linear Model analysis confirmed a relationship between abatacept exposure and impaired production of CXCL9, CXCL10, and IFN-γ from stimulated T cells. Cluster analysis confirms that cluster 1 (including abatacept and half of rituximab treated cases) showed a reduced IFN-γ response, as well as reduced monocyte-derived chemokines All groups of patients demonstrated the ability to generate specific CD4 T activated cells after spike proteins stimulation. After the third dose of vaccine, abatacept-treated patients acquired the ability to produce a strong antibody response, showing an anti-S titer significantly higher compared to that obtained after the second dose (p = 0.0047), and comparable with the anti-S titer of the other groups. CONCLUSIONS Patients treated with abatacept showed an impaired humoral immune response to two doses of COVID-19 vaccine. The third vaccine dose has been demonstrated to be useful to induce a more robust antibody response to balance an impaired T cell-mediated one. All patients, exposed to different immunosuppressive drugs, were able to produce specific CD4-activated T cells, after spike proteins stimulation. TRIAL REGISTRATION Local Ethical Committee NP4187.
Collapse
Affiliation(s)
- Federica Filippini
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Mauro Giacomelli
- Section of Microbiology, ASST Spedali Civili of Brescia, 25123, Brescia, Italy
| | - Chiara Bazzani
- Rheumatology and Clinical Immunology, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Micaela Fredi
- Rheumatology and Clinical Immunology, ASST Spedali Civili of Brescia and Department of Clinical and Experimental Sciences, University of Brescia, 25123, Brescia, Italy
| | - Paolo Semeraro
- Rheumatology and Clinical Immunology, ASST Spedali Civili of Brescia and Department of Clinical and Experimental Sciences, University of Brescia, 25123, Brescia, Italy
| | - Cesare Tomasi
- Rheumatology and Clinical Immunology, ASST Spedali Civili of Brescia and Department of Clinical and Experimental Sciences, University of Brescia, 25123, Brescia, Italy
| | - Franco Franceschini
- Rheumatology and Clinical Immunology, ASST Spedali Civili of Brescia and Department of Clinical and Experimental Sciences, University of Brescia, 25123, Brescia, Italy
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Ilaria Cavazzana
- Rheumatology and Clinical Immunology, ASST Spedali Civili of Brescia, Brescia, Italy.
| | - Cinzia Giagulli
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| |
Collapse
|
22
|
Mohammed H Al-Mquter LF, Abdul Azeez Atiayh S. Evaluation of IL-6, IL-25 & IL-35 in the COVID 19 Patients and their Correlation to Demography Data in the Symptomatic Patients. ARCHIVES OF RAZI INSTITUTE 2023; 78:1049-1056. [PMID: 38028847 PMCID: PMC10657937 DOI: 10.22092/ari.2022.360087.2547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/26/2022] [Indexed: 12/01/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2, SARS-CoV-2, was first discovered in Wuhan, Hubei province, China. Cytokines play a critical role in COVID-19 infections through their inflammatory or anti-inflammatory activities. This study aimed to detect the diagnostic value of and the relationship between the interleukins under study, in addition to their relationship with demographic data in COVID-19 patients. Patients with a confirmed diagnosis of COVID-19 based on laboratory (PCR) results and the healthy control group were given their section of this investigation. The patient group had 120 COVID-19 patients, including 62 males and 58 females, while the control group consisted of 32 individuals (22 males and 10 females). The subdivision was then performed according to their vaccination status, chronic diseases, gender, and residence. Cytokine levels were detected using the ELISA technique. The immunological status of COVID-19 patients was determined by measuring interleukin (IL)-6, IL-25, and IL-35. During the research, it was found that IL-6 was highly significant in COVID-19 patients (0.001). However, its level was not significantly different (0.376) in patients regarding the type of chronic diseases, residence (0.353), and gender (0.574), but it was significantly different in vaccinated patients (0.029). It was also found that IL-6 is significantly correlated with IL-25 and IL-35. IL-25 was highly significant in COVID-19 patients (0.007), and there was a significant difference in its level in patients regarding the type of chronic disease (0.049). While there was no difference in terms of residence (0.421) and gender (0.681), corona vaccination showed a significant difference (0.047). IL-25 also had a significant correlation with IL-6 and IL-35. As for IL-35, it was significant in patients with COVID-19 (0.013) but not significantly different regarding chronic diseases (0.344), residence (0.877), or gender (0.800). However, it was significantly different in vaccinated patients, compared to the non-vaccinated ones. IL-35 was found to be significantly correlated with IL-25 and IL-6 (0.000). The examined interleukins increased in COVID-19 individuals. IL-6 remains an excellent marker for determining the immune state of patients with COVID-19. There were also strong correlations between the interleukins under study in COVID-19 patients. However, there was no relationship between age, residence, gender, and the concentration of studied cytokines. IL-25 increases significantly in COVID-19 patients suffering from chronic diseases. Therefore, it is more efficient in the follow-up of patients.
Collapse
Affiliation(s)
| | - S Abdul Azeez Atiayh
- Microbiology Department, Cancer Research Unit, College of Medicine, University of Thi-Qar, Nasiriyah, Iraq
| |
Collapse
|
23
|
Tartof SY, Xie F, Yadav R, Wernli KJ, Martin ET, Belongia EA, Gaglani M, Zimmerman RK, Talbot HK, Thornburg N, Flannery B, US Flu VE Network Investigators. Prior SARS-CoV-2 infection and COVID-19 vaccine effectiveness against outpatient illness during widespread circulation of SARS-CoV-2 Omicron variant, US Flu VE network. Influenza Other Respir Viruses 2023; 17:e13143. [PMID: 37246146 PMCID: PMC10209645 DOI: 10.1111/irv.13143] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND We estimated combined protection conferred by prior SARS-CoV-2 infection and COVID-19 vaccination against COVID-19-associated acute respiratory illness (ARI). METHODS During SARS-CoV-2 Delta (B.1.617.2) and Omicron (B.1.1.529) variant circulation between October 2021 and April 2022, prospectively enrolled adult patients with outpatient ARI had respiratory and filter paper blood specimens collected for SARS-CoV-2 molecular testing and serology. Dried blood spots were tested for immunoglobulin-G antibodies against SARS-CoV-2 nucleocapsid (NP) and spike protein receptor binding domain antigen using a validated multiplex bead assay. Evidence of prior SARS-CoV-2 infection also included documented or self-reported laboratory-confirmed COVID-19. We used documented COVID-19 vaccination status to estimate vaccine effectiveness (VE) by multivariable logistic regression by prior infection status. RESULTS Four hundred fifty-five (29%) of 1577 participants tested positive for SARS-CoV-2 infection at enrollment; 209 (46%) case-patients and 637 (57%) test-negative patients were NP seropositive, had documented previous laboratory-confirmed COVID-19, or self-reported prior infection. Among previously uninfected patients, three-dose VE was 97% (95% confidence interval [CI], 60%-99%) against Delta, but not statistically significant against Omicron. Among previously infected patients, three-dose VE was 57% (CI, 20%-76%) against Omicron; VE against Delta could not be estimated. CONCLUSIONS Three mRNA COVID-19 vaccine doses provided additional protection against SARS-CoV-2 Omicron variant-associated illness among previously infected participants.
Collapse
Affiliation(s)
- Sara Y. Tartof
- Department of Research and EvaluationKaiser Permanente Southern CaliforniaPasadenaCaliforniaUSA
- Department of Health Systems ScienceKaiser Permanente Bernard J. Tyson School of MedicinePasadenaCaliforniaUSA
| | - Fagen Xie
- Department of Research and EvaluationKaiser Permanente Southern CaliforniaPasadenaCaliforniaUSA
| | - Ruchi Yadav
- Centers for Disease Control and PreventionAtlantaGeorgiaUSA
| | - Karen J. Wernli
- Kaiser Permanente Washington Health Research InstituteSeattleWashingtonUSA
| | - Emily T. Martin
- University of Michigan School of Public HealthAnn ArborMichiganUSA
| | | | - Manjusha Gaglani
- Baylor Scott & White HealthTempleTexasUSA
- Texas A&M University College of MedicineTempleTexasUSA
| | | | | | | | | | | |
Collapse
|
24
|
Pozdnyakova V, Weber B, Cheng S, Ebinger JE. Review of Immunologic Manifestations of COVID-19 Infection and Vaccination. Heart Fail Clin 2023; 19:177-184. [PMID: 36863809 PMCID: PMC9973544 DOI: 10.1016/j.hfc.2022.08.006] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
We herein summarize currently available and clinically relevant information regarding the human immune responses to SARS-CoV-2 infection and vaccination, in relation to COVID-19 outcomes with a focus on acute respiratory distress syndrome (ARDS) and myocarditis.
Collapse
Affiliation(s)
- Valeriya Pozdnyakova
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, D4005, Los Angeles, CA 90048, USA
| | - Brittany Weber
- Carl J. and Ruth Shapiro Cardiovascular Center, Brigham and Women's Hospital, 70 Francis Street, Boston, MA 02115, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South Vicente Boulevard, Suite A3100, Los Angeles, CA 90048, USA
| | - Joseph E Ebinger
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South Vicente Boulevard, Suite A3100, Los Angeles, CA 90048, USA.
| |
Collapse
|
25
|
Stein C, Nassereldine H, Sorensen RJD, Amlag JO, Bisignano C, Byrne S, Castro E, Coberly K, Collins JK, Dalos J, Daoud F, Deen A, Gakidou E, Giles JR, Hulland EN, Huntley BM, Kinzel KE, Lozano R, Mokdad AH, Pham T, Pigott DM, Reiner Jr. RC, Vos T, Hay SI, Murray CJL, Lim SS. Past SARS-CoV-2 infection protection against re-infection: a systematic review and meta-analysis. Lancet 2023; 401:833-842. [PMID: 36930674 PMCID: PMC9998097 DOI: 10.1016/s0140-6736(22)02465-5] [Citation(s) in RCA: 213] [Impact Index Per Article: 106.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 02/18/2023]
Abstract
BACKGROUND Understanding the level and characteristics of protection from past SARS-CoV-2 infection against subsequent re-infection, symptomatic COVID-19 disease, and severe disease is essential for predicting future potential disease burden, for designing policies that restrict travel or access to venues where there is a high risk of transmission, and for informing choices about when to receive vaccine doses. We aimed to systematically synthesise studies to estimate protection from past infection by variant, and where data allow, by time since infection. METHODS In this systematic review and meta-analysis, we identified, reviewed, and extracted from the scientific literature retrospective and prospective cohort studies and test-negative case-control studies published from inception up to Sept 31, 2022, that estimated the reduction in risk of COVID-19 among individuals with a past SARS-CoV-2 infection in comparison to those without a previous infection. We meta-analysed the effectiveness of past infection by outcome (infection, symptomatic disease, and severe disease), variant, and time since infection. We ran a Bayesian meta-regression to estimate the pooled estimates of protection. Risk-of-bias assessment was evaluated using the National Institutes of Health quality-assessment tools. The systematic review was PRISMA compliant and was registered with PROSPERO (number CRD42022303850). FINDINGS We identified a total of 65 studies from 19 different countries. Our meta-analyses showed that protection from past infection and any symptomatic disease was high for ancestral, alpha, beta, and delta variants, but was substantially lower for the omicron BA.1 variant. Pooled effectiveness against re-infection by the omicron BA.1 variant was 45·3% (95% uncertainty interval [UI] 17·3-76·1) and 44·0% (26·5-65·0) against omicron BA.1 symptomatic disease. Mean pooled effectiveness was greater than 78% against severe disease (hospitalisation and death) for all variants, including omicron BA.1. Protection from re-infection from ancestral, alpha, and delta variants declined over time but remained at 78·6% (49·8-93·6) at 40 weeks. Protection against re-infection by the omicron BA.1 variant declined more rapidly and was estimated at 36·1% (24·4-51·3) at 40 weeks. On the other hand, protection against severe disease remained high for all variants, with 90·2% (69·7-97·5) for ancestral, alpha, and delta variants, and 88·9% (84·7-90·9) for omicron BA.1 at 40 weeks. INTERPRETATION Protection from past infection against re-infection from pre-omicron variants was very high and remained high even after 40 weeks. Protection was substantially lower for the omicron BA.1 variant and declined more rapidly over time than protection against previous variants. Protection from severe disease was high for all variants. The immunity conferred by past infection should be weighed alongside protection from vaccination when assessing future disease burden from COVID-19, providing guidance on when individuals should be vaccinated, and designing policies that mandate vaccination for workers or restrict access, on the basis of immune status, to settings where the risk of transmission is high, such as travel and high-occupancy indoor settings. FUNDING Bill & Melinda Gates Foundation, J Stanton, T Gillespie, and J and E Nordstrom.
Collapse
|
26
|
Humoral and cellular response after BNT162b2 vaccine booster in hemodialysis patients and kidney transplant recipients. Clin Exp Nephrol 2023; 27:445-453. [PMID: 36795176 PMCID: PMC9933795 DOI: 10.1007/s10157-023-02317-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 01/10/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND Vulnerable populations, such as hemodialysis (HD) patients and kidney transplant (RTx) recipients, have priority for anti-COVID-19 vaccination, because of their impaired immune status. Here, we investigated the immune response after vaccination with BNT162b2 (two doses plus booster) in HD and RTx patients. METHODS A prospective, observational study was started in two homogeneous groups of 55 HD and 51 RTx patients previously matched from a cohort of 336 patients. Anti-RBD IgG levels, assayed after the second dose with BNT162b2 mRNA, were used to stratify subjects into quintiles. After the second dose and after booster, anti-RBD and IGRA test were evaluated in RTx and HD, belonging to the first and fifth quintiles. RESULTS After the second dose of vaccine, the median circulating levels of anti-RBD IgG were significantly higher in HD (1456 AU/mL) compared to RTx (27.30 AU/mL). IGRA test showed significantly higher values in the HD (382 mIU/mL) compared with the RTx (73 mIU/mL). After the booster, humoral response increased significantly in both HD (p = 0.0002) and RTx groups (p = 0.009), whereas the T-cellular immunity remained essentially stable in most patients. In RTx patients with a low humoral response after the second dose, the third dose did not significantly strengthen either humoral or cellular immunity. CONCLUSIONS For HD and RTx, there is great variability in the humoral response to anti-COVID-19 vaccination, with a stronger response in the HD group. The booster dose was ineffective at reinforcing the humoral and cellular immune response in most RTx patients hyporesponsive to the second dose.
Collapse
|
27
|
Gupta M, Balachandran H, Louie RHY, Li H, Agapiou D, Keoshkerian E, Christ D, Rawlinson W, Mina MM, Post JJ, Hudson B, Gilroy N, Konecny P, Bartlett AW, Sasson SC, Ahlenstiel G, Dwyer D, Lloyd AR, Martinello M, Luciani F, Bull RA, the COSIN study group. High activation levels maintained in receptor-binding domain-specific memory B cells in people with severe coronavirus disease 2019. Immunol Cell Biol 2023; 101:142-155. [PMID: 36353774 PMCID: PMC9878167 DOI: 10.1111/imcb.12607] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 09/02/2022] [Accepted: 11/09/2022] [Indexed: 11/11/2022]
Abstract
The long-term health consequences of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are still being understood. The molecular and phenotypic properties of SARS-CoV-2 antigen-specific T cells suggest a dysfunctional profile that persists in convalescence in those who were severely ill. By contrast, the antigen-specific memory B-cell (MBC) population has not yet been analyzed to the same degree, but phenotypic analysis suggests differences following recovery from mild or severe coronavirus disease 2019 (COVID-19). Here, we performed single-cell molecular analysis of the SARS-CoV-2 receptor-binding domain (RBD)-specific MBC population in three patients after severe COVID-19 and four patients after mild/moderate COVID-19. We analyzed the transcriptomic and B-cell receptor repertoire profiles at ~2 months and ~4 months after symptom onset. Transcriptomic analysis revealed a higher level of tumor necrosis factor-alpha (TNF-α) signaling via nuclear factor-kappa B in the severe group, involving CD80, FOS, CD83 and TNFAIP3 genes that was maintained over time. We demonstrated the presence of two distinct activated MBCs subsets based on expression of CD80hi TNFAIP3hi and CD11chi CD95hi at the transcriptome level. Both groups revealed an increase in somatic hypermutation over time, indicating progressive evolution of humoral memory. This study revealed distinct molecular signatures of long-term RBD-specific MBCs in convalescence, indicating that the longevity of these cells may differ depending on acute COVID-19 severity.
Collapse
Affiliation(s)
- Money Gupta
- Faculty of Medicine, School of Medical SciencesUniversity of New South Wales AustraliaSydneyNSWAustralia
- The Kirby Institute, University of New South Wales, AustraliaSydneyNSWAustralia
| | - Harikrishnan Balachandran
- Faculty of Medicine, School of Medical SciencesUniversity of New South Wales AustraliaSydneyNSWAustralia
- The Kirby Institute, University of New South Wales, AustraliaSydneyNSWAustralia
| | - Raymond H Y Louie
- Faculty of Medicine, School of Medical SciencesUniversity of New South Wales AustraliaSydneyNSWAustralia
- The Kirby Institute, University of New South Wales, AustraliaSydneyNSWAustralia
| | - Hui Li
- The Kirby Institute, University of New South Wales, AustraliaSydneyNSWAustralia
| | - David Agapiou
- The Kirby Institute, University of New South Wales, AustraliaSydneyNSWAustralia
| | | | - Daniel Christ
- Antibody Therapeutics LabGarvan Institute of Medical ResearchDarlinghurstNSWAustralia
| | - William Rawlinson
- Faculty of Medicine, School of Medical SciencesUniversity of New South Wales AustraliaSydneyNSWAustralia
- Serology and Virology Division, Department of MicrobiologyNSW Health Pathology, Prince of Wales HospitalSydneyNSWAustralia
| | | | - Jeffrey J Post
- Prince of Wales Clinical SchoolUniversity of New South Wales, AustraliaSydneyNSWAustralia
| | - Bernard Hudson
- Infectious diseasesRoyal North Shore HospitalSydneyNSWAustralia
| | - Nicky Gilroy
- Infectious DiseasesWestmead HospitalSydneyNSWAustralia
| | - Pamela Konecny
- St George and Sutherland Clinical SchoolUniversity of New South Wales, SydneySydneyNSWAustralia
| | - Adam W Bartlett
- Faculty of Medicine, School of Medical SciencesUniversity of New South Wales AustraliaSydneyNSWAustralia
- The Kirby Institute, University of New South Wales, AustraliaSydneyNSWAustralia
- Sydney Children's Hospital RandwickSydneyNSWAustralia
| | - Sarah C Sasson
- The Kirby Institute, University of New South Wales, AustraliaSydneyNSWAustralia
| | | | - Dominic Dwyer
- Infectious DiseasesWestmead HospitalSydneyNSWAustralia
| | - Andrew R Lloyd
- The Kirby Institute, University of New South Wales, AustraliaSydneyNSWAustralia
| | - Marianne Martinello
- The Kirby Institute, University of New South Wales, AustraliaSydneyNSWAustralia
- Infectious DiseasesWestmead HospitalSydneyNSWAustralia
- Blacktown Mount Druitt HospitalBlacktownNSWAustralia
| | - Fabio Luciani
- Faculty of Medicine, School of Medical SciencesUniversity of New South Wales AustraliaSydneyNSWAustralia
- The Kirby Institute, University of New South Wales, AustraliaSydneyNSWAustralia
| | - Rowena A Bull
- Faculty of Medicine, School of Medical SciencesUniversity of New South Wales AustraliaSydneyNSWAustralia
- The Kirby Institute, University of New South Wales, AustraliaSydneyNSWAustralia
| | | |
Collapse
|
28
|
Ruiz Ortega M, Spisak N, Mora T, Walczak AM. Modeling and predicting the overlap of B- and T-cell receptor repertoires in healthy and SARS-CoV-2 infected individuals. PLoS Genet 2023; 19:e1010652. [PMID: 36827454 PMCID: PMC10075420 DOI: 10.1371/journal.pgen.1010652] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/05/2023] [Accepted: 02/02/2023] [Indexed: 02/26/2023] Open
Abstract
Adaptive immunity's success relies on the extraordinary diversity of protein receptors on B and T cell membranes. Despite this diversity, the existence of public receptors shared by many individuals gives hope for developing population-wide vaccines and therapeutics. Using probabilistic modeling, we show many of these public receptors are shared by chance in healthy individuals. This predictable overlap is driven not only by biases in the random generation process of receptors, as previously reported, but also by their common functional selection. However, the model underestimates sharing between repertoires of individuals infected with SARS-CoV-2, suggesting strong specific antigen-driven convergent selection. We exploit this discrepancy to identify COVID-associated receptors, which we validate against datasets of receptors with known viral specificity. We study their properties in terms of sequence features and network organization, and use them to design an accurate diagnostic tool for predicting SARS-CoV-2 status from repertoire data.
Collapse
Affiliation(s)
- María Ruiz Ortega
- Laboratoire de physique de l’École Normale Supérieure, CNRS, PSL University, Sorbonne Université, and Université de Paris, Paris, France
| | - Natanael Spisak
- Laboratoire de physique de l’École Normale Supérieure, CNRS, PSL University, Sorbonne Université, and Université de Paris, Paris, France
| | - Thierry Mora
- Laboratoire de physique de l’École Normale Supérieure, CNRS, PSL University, Sorbonne Université, and Université de Paris, Paris, France
| | - Aleksandra M. Walczak
- Laboratoire de physique de l’École Normale Supérieure, CNRS, PSL University, Sorbonne Université, and Université de Paris, Paris, France
| |
Collapse
|
29
|
Somogyi E, Kremlitzka M, Csiszovszki Z, Molnár L, Lőrincz O, Tóth J, de Waal L, Pattijn S, Reineking W, Beineke A, Tőke ER. T cell immunity ameliorates COVID-19 disease severity and provides post-exposure prophylaxis after peptide-vaccination, in Syrian hamsters. Front Immunol 2023; 14:1111629. [PMID: 36761759 PMCID: PMC9902696 DOI: 10.3389/fimmu.2023.1111629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/05/2023] [Indexed: 01/25/2023] Open
Abstract
Background The emergence of novel SARS-CoV-2 variants that resist neutralizing antibodies drew the attention to cellular immunity and calls for the development of alternative vaccination strategies to combat the pandemic. Here, we have assessed the kinetics of T cell responses and protective efficacy against severe COVID-19 in pre- and post-exposure settings, elicited by PolyPEPI-SCoV-2, a peptide based T cell vaccine. Methods 75 Syrian hamsters were immunized subcutaneously with PolyPEPI-SCoV-2 on D0 and D14. On D42, hamsters were intranasally challenged with 102 TCID50 of the virus. To analyze immunogenicity by IFN-γ ELISPOT and antibody secretion, lymphoid tissues were collected both before (D0, D14, D28, D42) and after challenge (D44, D46, D49). To measure vaccine efficacy, lung tissue, throat swabs and nasal turbinate samples were assessed for viral load and histopathological changes. Further, body weight was monitored on D0, D28, D42 and every day after challenge. Results The vaccine induced robust activation of T cells against all SARS-CoV-2 structural proteins that were rapidly boosted after virus challenge compared to control animals (~4-fold, p<0.05). A single dose of PolyPEPI-SCoV-2 administered one day after challenge also resulted in elevated T cell response (p<0.01). The vaccination did not induce virus-specific antibodies and viral load reduction. Still, peptide vaccination significantly reduced body weight loss (p<0.001), relative lung weight (p<0.05) and lung lesions (p<0.05), in both settings. Conclusion Our study provides first proof of concept data on the contribution of T cell immunity on disease course and provide rationale for the use of T cell-based peptide vaccines against both novel SARS-CoV-2 variants and supports post-exposure prophylaxis as alternative vaccination strategy against COVID-19.
Collapse
Affiliation(s)
- Eszter Somogyi
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
- PepTC Vaccines Ltd, London, United Kingdom
| | - Mariann Kremlitzka
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
- PepTC Vaccines Ltd, London, United Kingdom
| | - Zsolt Csiszovszki
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
- PepTC Vaccines Ltd, London, United Kingdom
| | - Levente Molnár
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
- PepTC Vaccines Ltd, London, United Kingdom
| | - Orsolya Lőrincz
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
- PepTC Vaccines Ltd, London, United Kingdom
| | - József Tóth
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
- PepTC Vaccines Ltd, London, United Kingdom
| | - Leon de Waal
- Viroclinics Biosciences B.V., Viroclinics Xplore, Schaijk, Netherlands
| | - Sofie Pattijn
- ImmunXperts Société Anonyme, Q2 Solutions Company, Gosselies, Belgium
| | - Wencke Reineking
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Enikő R. Tőke
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
- PepTC Vaccines Ltd, London, United Kingdom
| |
Collapse
|
30
|
Tartof SY, Xie F, Yadav R, Wernli KJ, Martin ET, Belongia EA, Gaglani M, Zimmerman RK, Talbot HK, Thornburg N, Flannery B. Prior SARS-CoV-2 Infection and COVID-19 Vaccine Effectiveness against Outpatient Illness during Widespread Circulation of SARS-CoV-2 Omicron Variant, US Flu VE Network. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.10.23284397. [PMID: 36711929 PMCID: PMC9882409 DOI: 10.1101/2023.01.10.23284397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Background We estimated combined protection conferred by prior SARS-CoV-2 infection and COVID-19 vaccination against COVID-19-associated acute respiratory illness (ARI). Methods During SARS-CoV-2 Delta (B.1.617.2) and Omicron (B.1.1.529) variant circulation between October 2021 and April 2022, prospectively enrolled adult patients with outpatient ARI had respiratory and filter paper blood specimens collected for SARS-CoV-2 molecular testing and serology. Dried blood spots were tested for immunoglobulin-G antibodies against SARS-CoV-2 nucleocapsid (NP) and spike protein receptor binding domain antigen using a validated multiplex bead assay. Evidence of prior SARS-CoV-2 infection also included documented or self-reported laboratory-confirmed COVID-19. We used documented COVID-19 vaccination status to estimate vaccine effectiveness (VE) by multivariable logistic regression by prior infection status. Results 455 (29%) of 1577 participants tested positive for SARS-CoV-2 infection at enrollment; 209 (46%) case-patients and 637 (57%) test-negative patients were NP seropositive, had documented previous laboratory-confirmed COVID-19, or self-reported prior infection. Among previously uninfected patients, three-dose VE was 97% (95% confidence interval [CI], 60%- 99%) against Delta, but not statistically significant against Omicron. Among previously infected patients, three-dose VE was 57% (CI, 20%-76%) against Omicron; VE against Delta could not be estimated. Conclusions Three mRNA COVID-19 vaccine doses provided additional protection against SARS-CoV-2 Omicron variant-associated illness among previously infected participants.
Collapse
Affiliation(s)
- Sara Y Tartof
- Kaiser Permanente Southern California, Department of Research & Evaluation
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA USA
| | - Fagen Xie
- Kaiser Permanente Southern California, Department of Research & Evaluation
| | - Ruchi Yadav
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Karen J Wernli
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Emily T Martin
- University of Michigan School of Public Health, Ann Arbor, MI, USA
| | | | - Manjusha Gaglani
- Baylor Scott & White Health, Temple, TX, USA
- Texas A&M University College of Medicine, Temple, TX, USA
| | | | - H Keipp Talbot
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | |
Collapse
|
31
|
Rümke LW, Smit WL, Bossink A, Limonard GJM, Muilwijk D, Haas LEM, Reusken C, van der Wal S, Thio BJ, van Os YMG, Gremmels H, Beekman JM, Nijhuis M, Wensing AMJ, Heron M, Thijsen SFT. Impaired SARS-CoV-2 specific T-cell response in patients with severe COVID-19. Front Immunol 2023; 14:1046639. [PMID: 37168853 PMCID: PMC10165493 DOI: 10.3389/fimmu.2023.1046639] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 03/28/2023] [Indexed: 05/13/2023] Open
Abstract
Cellular immune responses are of pivotal importance to understand SARS-CoV-2 pathogenicity. Using an enzyme-linked immunosorbent spot (ELISpot) interferon-γ release assay with wild-type spike, membrane and nucleocapsid peptide pools, we longitudinally characterized functional SARS-CoV-2 specific T-cell responses in a cohort of patients with mild, moderate and severe COVID-19. All patients were included before emergence of the Omicron (B.1.1.529) variant. Our most important finding was an impaired development of early IFN-γ-secreting virus-specific T-cells in severe patients compared to patients with moderate disease, indicating that absence of virus-specific cellular responses in the acute phase may act as a prognostic factor for severe disease. Remarkably, in addition to reactivity against the spike protein, a substantial proportion of the SARS-CoV-2 specific T-cell response was directed against the conserved membrane protein. This may be relevant for diagnostics and vaccine design, especially considering new variants with heavily mutated spike proteins. Our data further strengthen the hypothesis that dysregulated adaptive immunity plays a central role in COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
- Lidewij W. Rümke
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
- Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Wouter L. Smit
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
- Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ailko Bossink
- Department of Pulmonary Diseases, Diakonessenhuis Utrecht, Utrecht, Netherlands
| | - Gijs J. M. Limonard
- Department of Pulmonary Diseases, Diakonessenhuis Utrecht, Utrecht, Netherlands
| | - Danya Muilwijk
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center, Utrecht University, Utrecht, Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center, Utrecht University, Utrecht, Netherlands
| | - Lenneke E. M. Haas
- Department of Intensive Care, Diakonessenhuis Utrecht, Utrecht, Netherlands
| | - Chantal Reusken
- Centre for Infectious Disease Control, WHO Reference Laboratory for COVID-19, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Sanne van der Wal
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
| | - Bing J. Thio
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
| | - Yvonne M. G. van Os
- Occupational Health Office, Department of Human Resources, University Medical Center Utrecht, Utrecht, Netherlands
| | - Hendrik Gremmels
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
- Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jeffrey M. Beekman
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center, Utrecht University, Utrecht, Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center, Utrecht University, Utrecht, Netherlands
| | - Monique Nijhuis
- Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Annemarie M. J. Wensing
- Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Michiel Heron
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
- *Correspondence: Michiel Heron,
| | - Steven F. T. Thijsen
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
| |
Collapse
|
32
|
Petersen MS, Pérez-Alós L, Armenteros JJA, Hansen CB, Fjallsbak JP, Larsen S, Hansen JL, Jarlhelt I, Kristiansen MF, Við Streym F, Á Steig B, Christiansen DH, Møller LF, Strøm M, Andorsdóttir G, Gaini S, Weihe P, Garred P. Factors influencing the immune response over 15 months after SARS-CoV-2 infection: A longitudinal population-wide study in the Faroe Islands. J Intern Med 2023; 293:63-81. [PMID: 35996938 PMCID: PMC9539344 DOI: 10.1111/joim.13560] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND The durability of SARS-CoV-2 antibody response and the resulting immunity to COVID-19 is unclear. OBJECTIVES To investigate long-term humoral immunity to SARS-CoV-2. METHODS In this nationwide, longitudinal study, we determined antibody response in 411 patients aged 0-93 years from two waves of infections (March to December 2020) contributing 1063 blood samples. Each individual had blood drawn on 4-5 occasions 1-15 months after disease onset. We measured total anti-SARS-CoV-2 receptor-binding domain (RBD) antibody using a qualitative RBD sandwich ELISA, IgM, IgG and IgA levels using an quantitative in-house ELISA-based assay and neutralizing antibodies (NAbs) using an in-house ELISA-based pseudoneutralizing assay. IgG subclasses were analyzed in a subset of samples by ELISA-based assay. We used nonlinear models to study the durability of SARS-CoV-2 antibody responses and its influence over time. RESULTS After 15 months, 94% still had detectable circulating antibodies, mainly the IgG isotype, and 92% had detectable NAbs. The distribution of IgG antibodies varied significantly over time, characterized by a biphasic pattern with an initial decline followed by a plateau after approximately 7 months. However, the NAbs remained relatively stable throughout the period. The strength of the antibody response was influenced by smoking and hospitalization, with lower IgG levels in smokers and higher levels in hospitalized individuals. Antibody stability over time was mainly associated with male sex and older age with higher initial levels but more marked decrease. CONCLUSIONS The humoral immune response to SARS-CoV-2 infection varies depending on behavioral factors and disease severity, and antibody stability over 15 months was associated with sex and age.
Collapse
Affiliation(s)
- Maria Skaalum Petersen
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands.,Centre of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jose Juan A Armenteros
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Cecilie B Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Sólrun Larsen
- Faroese Food and Veterinary Authority, Tórshavn, Faroe Islands
| | | | - Ida Jarlhelt
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marnar F Kristiansen
- Centre of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands.,COVID-19 Task Force, Ministry of Health, Tórshavn, Faroe Islands.,Medical Department, National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
| | - Fríða Við Streym
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands
| | - Bjarni Á Steig
- COVID-19 Task Force, Ministry of Health, Tórshavn, Faroe Islands.,Medical Department, National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
| | | | | | - Marin Strøm
- Centre of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands.,Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | | | - Shahin Gaini
- Centre of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands.,Medical Department, National Hospital of the Faroe Islands, Tórshavn, Faroe Islands.,Department of Infectious Diseases, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Pál Weihe
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands.,Centre of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Tu W, Zhang P, Roberts A, Allen KS, Williams J, Embi P, Grannis S. SARS-CoV-2 Infection, Hospitalization, and Death in Vaccinated and Infected Individuals by Age Groups in Indiana, 2021‒2022. Am J Public Health 2023; 113:96-104. [PMID: 36516380 PMCID: PMC9755951 DOI: 10.2105/ajph.2022.307112] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 12/15/2022]
Abstract
Objectives. To assess the effectiveness of vaccine-induced immunity against new infections, all-cause emergency department (ED) and hospital visits, and mortality in Indiana. Methods. Combining statewide testing and immunization data with patient medical records, we matched individuals who received at least 1 dose of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines with individuals with previous SARS-CoV-2 infection on index date, age, gender, race/ethnicity, zip code, and clinical diagnoses. We compared the cumulative incidence of infection, all-cause ED visits, hospitalizations, and mortality. Results. We matched 267 847 pairs of individuals. Six months after the index date, the incidence of SARS-CoV-2 infection was significantly higher in vaccine recipients (6.7%) than the previously infected (2.9%). All-cause mortality in the vaccinated, however, was 37% lower than that of the previously infected. The rates of all-cause ED visits and hospitalizations were 24% and 37% lower in the vaccinated than in the previously infected. Conclusions. The significantly lower rates of all-cause ED visits, hospitalizations, and mortality in the vaccinated highlight the real-world benefits of vaccination. The data raise questions about the wisdom of reliance on natural immunity when safe and effective vaccines are available. (Am J Public Health. 2023;113(1):96-104. https://doi.org/10.2105/AJPH.2022.307112).
Collapse
Affiliation(s)
- Wanzhu Tu
- Wanzhu Tu and Pengyue Zhang are with the Indiana University School of Medicine and the Fairbanks School of Public Health, Indianapolis. Peter Embi and Shaun Grannis are with the Indiana University School of Medicine, Indianapolis. Anna Roberts, Katie S. Allen, and Jennifer Williams are with the Regenstrief Institute Inc, Indianapolis
| | - Pengyue Zhang
- Wanzhu Tu and Pengyue Zhang are with the Indiana University School of Medicine and the Fairbanks School of Public Health, Indianapolis. Peter Embi and Shaun Grannis are with the Indiana University School of Medicine, Indianapolis. Anna Roberts, Katie S. Allen, and Jennifer Williams are with the Regenstrief Institute Inc, Indianapolis
| | - Anna Roberts
- Wanzhu Tu and Pengyue Zhang are with the Indiana University School of Medicine and the Fairbanks School of Public Health, Indianapolis. Peter Embi and Shaun Grannis are with the Indiana University School of Medicine, Indianapolis. Anna Roberts, Katie S. Allen, and Jennifer Williams are with the Regenstrief Institute Inc, Indianapolis
| | - Katie S Allen
- Wanzhu Tu and Pengyue Zhang are with the Indiana University School of Medicine and the Fairbanks School of Public Health, Indianapolis. Peter Embi and Shaun Grannis are with the Indiana University School of Medicine, Indianapolis. Anna Roberts, Katie S. Allen, and Jennifer Williams are with the Regenstrief Institute Inc, Indianapolis
| | - Jennifer Williams
- Wanzhu Tu and Pengyue Zhang are with the Indiana University School of Medicine and the Fairbanks School of Public Health, Indianapolis. Peter Embi and Shaun Grannis are with the Indiana University School of Medicine, Indianapolis. Anna Roberts, Katie S. Allen, and Jennifer Williams are with the Regenstrief Institute Inc, Indianapolis
| | - Peter Embi
- Wanzhu Tu and Pengyue Zhang are with the Indiana University School of Medicine and the Fairbanks School of Public Health, Indianapolis. Peter Embi and Shaun Grannis are with the Indiana University School of Medicine, Indianapolis. Anna Roberts, Katie S. Allen, and Jennifer Williams are with the Regenstrief Institute Inc, Indianapolis
| | - Shaun Grannis
- Wanzhu Tu and Pengyue Zhang are with the Indiana University School of Medicine and the Fairbanks School of Public Health, Indianapolis. Peter Embi and Shaun Grannis are with the Indiana University School of Medicine, Indianapolis. Anna Roberts, Katie S. Allen, and Jennifer Williams are with the Regenstrief Institute Inc, Indianapolis
| |
Collapse
|
34
|
Dhawan M, Rabaan AA, Fawarah MMA, Almuthree SA, Alsubki RA, Alfaraj AH, Mashraqi MM, Alshamrani SA, Abduljabbar WA, Alwashmi ASS, Ibrahim FA, Alsaleh AA, Khamis F, Alsalman J, Sharma M, Emran TB. Updated Insights into the T Cell-Mediated Immune Response against SARS-CoV-2: A Step towards Efficient and Reliable Vaccines. Vaccines (Basel) 2023; 11:101. [PMID: 36679947 PMCID: PMC9861463 DOI: 10.3390/vaccines11010101] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/14/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
The emergence of novel variants of SARS-CoV-2 and their abilities to evade the immune response elicited through presently available vaccination makes it essential to recognize the mechanisms through which SARS-CoV-2 interacts with the human immune response. It is essential not only to comprehend the infection mechanism of SARS-CoV-2 but also for the generation of effective and reliable vaccines against COVID-19. The effectiveness of the vaccine is supported by the adaptive immune response, which mainly consists of B and T cells, which play a critical role in deciding the prognosis of the COVID-19 disease. T cells are essential for reducing the viral load and containing the infection. A plethora of viral proteins can be recognized by T cells and provide a broad range of protection, especially amid the emergence of novel variants of SARS-CoV-2. However, the hyperactivation of the effector T cells and reduced number of lymphocytes have been found to be the key characteristics of the severe disease. Notably, excessive T cell activation may cause acute respiratory distress syndrome (ARDS) by producing unwarranted and excessive amounts of cytokines and chemokines. Nevertheless, it is still unknown how T-cell-mediated immune responses function in determining the prognosis of SARS-CoV-2 infection. Additionally, it is unknown how the functional perturbations in the T cells lead to the severe form of the disease and to reduced protection not only against SARS-CoV-2 but many other viral infections. Hence, an updated review has been developed to understand the involvement of T cells in the infection mechanism, which in turn determines the prognosis of the disease. Importantly, we have also focused on the T cells' exhaustion under certain conditions and how these functional perturbations can be modulated for an effective immune response against SARS-CoV-2. Additionally, a range of therapeutic strategies has been discussed that can elevate the T cell-mediated immune response either directly or indirectly.
Collapse
Affiliation(s)
- Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana 141004, Punjab, India
- Trafford College, Altrincham, Manchester WA14 5PQ, UK
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Mahmoud M. Al Fawarah
- Microbiology Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
| | - Souad A. Almuthree
- Department of Infectious Disease, King Abdullah Medical City, Makkah 43442, Saudi Arabia
| | - Roua A. Alsubki
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11362, Saudi Arabia
| | - Amal H. Alfaraj
- Pediatric Department, Abqaiq General Hospital, First Eastern Health Cluster, Abqaiq 33261, Saudi Arabia
| | - Mutaib M. Mashraqi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Saleh A. Alshamrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Wesam A. Abduljabbar
- Department of Medical Laboratory Sciences, Fakeeh College for Medical Science, Jeddah 21134, Saudi Arabia
| | - Ameen S. S. Alwashmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Fatimah Al Ibrahim
- Infectious Disease Division, Department of Internal Medicine, Dammam Medical Complex, Dammam 32245, Saudi Arabia
| | - Abdulmonem A. Alsaleh
- Clinical Laboratory Science Department, Mohammed Al-Mana College for Medical Sciences, Dammam 34222, Saudi Arabia
| | - Faryal Khamis
- Infection Diseases Unit, Department of Internal Medicine, Royal Hospital, Muscat 1331, Oman
| | - Jameela Alsalman
- Infection Disease Unit, Department of Internal Medicine, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 435, Bahrain
| | - Manish Sharma
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, Punjab, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| |
Collapse
|
35
|
Akhtar M, Basher SR, Nizam NN, Kamruzzaman M, Khaton F, Banna HA, Kaisar MH, Karmakar PC, Hakim A, Akter A, Ahmed T, Tauheed I, Islam S, Ahmmed F, Mahamud S, Hasnat MA, Sumon MA, Rashed A, Ghosh S, Calderwood SB, Harris JB, Charles RC, LaRocque RC, Ryan ET, Banu S, Shirin T, Chowdhury F, Bhuiyan TR, Qadri F. Longevity of memory B cells and antibodies, as well as the polarization of effector memory helper T cells, are associated with disease severity in patients with COVID-19 in Bangladesh. Front Immunol 2022; 13:1052374. [PMID: 36578502 PMCID: PMC9791541 DOI: 10.3389/fimmu.2022.1052374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
The longevity of immune responses induced by different degrees of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection provides information important to understanding protection against coronavirus disease 2019 (COVID-19). Here, we report the persistence of SARS-CoV-2 spike receptor-binding domain (RBD) specific antibodies and memory B cells recognizing this antigen in sequential samples from patients in Bangladesh with asymptomatic, mild, moderate and severe COVID-19 out to six months following infection. Since the development of long-lived memory B cells, as well as antibody production, is likely to be dependent on T helper (Th) cells, we also investigated the phenotypic changes of Th cells in COVID-19 patients over time following infection. Our results show that patients with moderate to severe COVID-19 mounted significant levels of IgG antibodies out to six months following infection, while patients with asymptomatic or mild disease had significant levels of IgG antibodies out to 3 months following infection, but these then fell more rapidly at 6 months than in patients with higher disease severity. Patients from all severity groups developed circulating memory B cells (MBCs) specific to SARS-CoV-2 spike RBD by 3 months following infection, and these persisted until the last timepoint measured at 6 months. A T helper cell response with an effector memory phenotype was observed following infection in all symptomatic patients, while patients with asymptomatic infection had no significant increases in effector Th1, Th2 and Th17 effector memory cell responses. Our results suggest that the strength and magnitude of antibody and memory B cells induced following SARS-CoV-2 infection depend on the severity of the disease. Polarization of the Th cell response, with an increase in Th effector memory cells, occurs in symptomatic patients by day 7 following infection, with increases seen in Th1, Th2, Th17 and follicular helper T cell subsets.
Collapse
Affiliation(s)
- Marjahan Akhtar
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Salima Raiyan Basher
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Nuder Nower Nizam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Mohammad Kamruzzaman
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Fatema Khaton
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Hasan Al Banna
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - M Hasanul Kaisar
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Polash Chandra Karmakar
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Al Hakim
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
- Department of Genetic Engineering and Biotechnology, Jagannath University, Dhaka, Bangladesh
| | - Afroza Akter
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Tasnuva Ahmed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Imam Tauheed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Shaumik Islam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Faisal Ahmmed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Shakil Mahamud
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Mohammad Abul Hasnat
- Department of Cardiology, Department of Oncology, Kurmitola General Hospital, Dhaka, Bangladesh
| | - Mostafa Aziz Sumon
- Department of Cardiology, Department of Oncology, Kurmitola General Hospital, Dhaka, Bangladesh
| | - Asif Rashed
- Department of Microbiology, Department of Medicine, Mugda Medical College and Hospital, Dhaka, Bangladesh
| | - Shuvro Ghosh
- Department of Microbiology, Department of Medicine, Mugda Medical College and Hospital, Dhaka, Bangladesh
| | - Stephen B. Calderwood
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, United States
| | - Jason B. Harris
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, United States
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Richelle C. Charles
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, United States
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, United States
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Regina C. LaRocque
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, United States
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Edward T. Ryan
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, United States
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, United States
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Sayera Banu
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Tahmina Shirin
- Institute of Epidemiology, Disease Control and Research (IEDCR), Dhaka, Bangladesh
| | - Fahima Chowdhury
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Taufiqur Rahman Bhuiyan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Firdausi Qadri
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| |
Collapse
|
36
|
Belayachi J, Obtel M, Mhayi A, Razine R, Abouqal R. Long term effectiveness of inactivated vaccine BBIBP-CorV (Vero Cells) against COVID-19 associated severe and critical hospitalization in Morocco. PLoS One 2022; 17:e0278546. [PMID: 36477077 PMCID: PMC9728886 DOI: 10.1371/journal.pone.0278546] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND We provide national estimates of the real-world Vaccine effectiveness (VE) based on nationally available surveillance data. The study aimed to estimate the effectiveness of the inactivated Covid-19 vaccine BBIBP-CorV (Vero Cells) Sinopharm vaccine currently deployed in Morocco against SARS- CoV-2 severe disease/ hospitalization" within 9 months after vaccination. METHODS We conducted a test-negative, case-control study among a population aged 18 years or older who were tested by rt-PCR for SARS-CoV-2 infection from February to October 2021 in Morocco. From the national laboratory COVID-19 database; we identified cases who were rt-PCR positive amongst severe and critical COVID-19 cases and controls who had a negative rt-PCR test for SARS-CoV-2. From the national vaccination register (NVR); individuals vaccinated with COVID-19 Vaccine (Vero Cell) and those unvaccinated were identified and included in the study. The linkage between databases was conducted for the study of Vaccination status based on the timing of the vaccine receipt relative to the SARS-CoV-2 rt-PCR test date. For each person, who tested positive for SARS-CoV-2, we identified a propensity score-matched control participant who was tested negative. We estimated vaccine effectiveness against SARS- CoV-2 severe disease/ hospitalization using conditional logistic regression. RESULTS Among 12884 persons who tested positive and 12885 propensity score-matched control participants, the median age was 62 years, 47.2% of whom were female. As a function of time after vaccination of second dose vaccination, vaccine effectiveness during the first month was 88% (95% CI, 84-91), 87% (95% CI: 83-90) during the second and third month, 75% (95% CI: 67-80) during the fourth month, 61% (95% CI: 54-67) during the fifth month, and 64% (95% CI: 59-69) beyond the sixth month. VE remained high and stable during the first three months in the two-age subgroup. In the fourth month, the VE in the older population aged 60 years and above (64%) was reduced by 20 points compared to VE in the younger population (84%). CONCLUSION A Sinopharm vaccine is highly protective against serious SARS-CoV-2 infection under real-world conditions. Protection remained high and stable during the first three months following the second dose and decreases slightly beyond the fourth month especially beyond 60 years.
Collapse
Affiliation(s)
- Jihane Belayachi
- Acute Medical Unit, Ibn Sina University Hospital, Rabat, Morocco
- Laboratory of Biostatistics, Clinical, and Epidemiological Research, Faculty of Medicine and Pharmacy, Department of Public Health, Mohamed V University in Rabat, Rabat, Morocco
| | - Majdouline Obtel
- Laboratory of Biostatistics, Clinical, and Epidemiological Research, Faculty of Medicine and Pharmacy, Department of Public Health, Mohamed V University in Rabat, Rabat, Morocco
- Laboratory of Community Health (Public Health, Preventive Medicine and Hygiene), Faculty of Medicine and Pharmacy, Department of Public Health, Mohamed V University in Rabat, Rabat, Morocco
| | - Abdelkader Mhayi
- Department of Informatics, Ministry of Health and Social Protection, Rabat, Morocco
| | - Rachid Razine
- Laboratory of Biostatistics, Clinical, and Epidemiological Research, Faculty of Medicine and Pharmacy, Department of Public Health, Mohamed V University in Rabat, Rabat, Morocco
- Laboratory of Community Health (Public Health, Preventive Medicine and Hygiene), Faculty of Medicine and Pharmacy, Department of Public Health, Mohamed V University in Rabat, Rabat, Morocco
| | - Redouane Abouqal
- Acute Medical Unit, Ibn Sina University Hospital, Rabat, Morocco
- Laboratory of Biostatistics, Clinical, and Epidemiological Research, Faculty of Medicine and Pharmacy, Department of Public Health, Mohamed V University in Rabat, Rabat, Morocco
- * E-mail:
| |
Collapse
|
37
|
Humoral Responses in the Omicron Era Following 3-Dose SARS-CoV-2 Vaccine Series in Kidney Transplant Recipients. Transplant Direct 2022; 9:e1401. [DOI: 10.1097/txd.0000000000001401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 12/13/2022] Open
|
38
|
Van Delen M, Janssens I, Dams A, Roosens L, Ogunjimi B, Berneman ZN, Derdelinckx J, Cools N. Tolerogenic Dendritic Cells Induce Apoptosis-Independent T Cell Hyporesponsiveness of SARS-CoV-2-Specific T Cells in an Antigen-Specific Manner. Int J Mol Sci 2022; 23:15201. [PMID: 36499533 PMCID: PMC9740551 DOI: 10.3390/ijms232315201] [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: 11/09/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Although the global pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still ongoing, there are currently no specific and highly efficient drugs for COVID-19 available, particularly in severe cases. Recent findings demonstrate that severe COVID-19 disease that requires hospitalization is associated with the hyperactivation of CD4+ and CD8+ T cell subsets. In this study, we aimed to counteract this high inflammatory state by inducing T-cell hyporesponsiveness in a SARS-CoV-2-specific manner using tolerogenic dendritic cells (tolDC). In vitro-activated SARS-CoV-2-specific T cells were isolated and stimulated with SARS-CoV-2 peptide-loaded monocyte-derived tolDC or with SARS-CoV-2 peptide-loaded conventional (conv) DC. We demonstrate a significant decrease in the number of interferon (IFN)-γ spot-forming cells when SARS-CoV-2-specific T cells were stimulated with tolDC as compared to stimulation with convDC. Importantly, this IFN-γ downmodulation in SARS-CoV-2-specific T cells was antigen-specific, since T cells retain their capacity to respond to an unrelated antigen and are not mediated by T cell deletion. Altogether, we have demonstrated that SARS-CoV-2 peptide-pulsed tolDC induces SARS-CoV-2-specific T cell hyporesponsiveness in an antigen-specific manner as compared to stimulation with SARS-CoV-2-specific convDC. These observations underline the clinical potential of tolDC to correct the immunological imbalance in the critically ill.
Collapse
Affiliation(s)
- Mats Van Delen
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Ibo Janssens
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Amber Dams
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Laurence Roosens
- Laboratory of Clinical Biology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Benson Ogunjimi
- Centre for Health Economics Research & Modeling Infectious Diseases (CHERMID), VAXINFECTIO, University of Antwerp, 2610 Antwerp, Belgium
- Department of Paediatrics, Antwerp University Hospital, 2650 Edegem, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), VAXINFECTIO, University of Antwerp, 2610 Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
| | - Zwi N. Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Judith Derdelinckx
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| |
Collapse
|
39
|
Mohapatra RK, El-Shall NA, Tiwari R, Nainu F, Kandi V, Sarangi AK, Mohammed TA, Desingu PA, Chakraborty C, Dhama K. Need of booster vaccine doses to counteract the emergence of SARS-CoV-2 variants in the context of the Omicron variant and increasing COVID-19 cases: An update. Hum Vaccin Immunother 2022; 18:2065824. [PMID: 35594528 PMCID: PMC9897647 DOI: 10.1080/21645515.2022.2065824] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/25/2022] [Accepted: 04/07/2022] [Indexed: 02/06/2023] Open
Abstract
The emergence of different variants of SARS-CoV-2, including the Omicron (B.1.1.529) variant in November 2021, has resulted in a continuous major health concern at a global scale. Presently, the Omicron variant has spread very rapidly worldwide within a short time period. As the most mutated variant of SARS-CoV-2, Omicron has instilled serious uncertainties on the effectiveness of humoral adaptive immunity generated by COVID-19 vaccination or an active viral infection as well as the protection provided by antibody-based immunotherapies. Amidst such high public health concerns, the need to carry out booster vaccination has been emphasized. Current evidence reveals the importance of incorporating booster vaccination using several vaccine platforms, such as viral vector- and mRNA-based vaccines, as well as other platforms that are under explorative investigations. Further research is being conducted to assess the effectiveness and durability of protection provided by booster COVID-19 vaccination against Omicron and other SARS-CoV-2 variants.
Collapse
Affiliation(s)
| | - Nahed A. El-Shall
- Department of Poultry and Fish Diseases, Faculty of Veterinary Medicine, Alexandria University, Edfina, El-Beheira, Egypt
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit DeenDayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, India
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Venkataramana Kandi
- Department of Microbiology, Prathima Institute of Medical Sciences, Karimnagar, India
| | - Ashish K. Sarangi
- Department of Chemistry, School of Applied Sciences, Centurion University of Technology and Management, Balangir, India
| | | | | | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| |
Collapse
|
40
|
Rajamanickam A, Nathella PK, Venkataraman A, Varadarjan P, Kannan S, Pandiarajan AN, Renji RM, Elavarasan E, Thimmaiah A, Sasidaran K, Krishnamoorthy N, Natarajan S, Ramaswamy G, Sundaram B, Putlibai S, Hissar S, Selladurai E, Uma Devi KR, Nutman TB, Babu S. Unique cellular immune signatures of multisystem inflammatory syndrome in children. PLoS Pathog 2022; 18:e1010915. [PMID: 36322537 PMCID: PMC9629618 DOI: 10.1371/journal.ppat.1010915] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 10/04/2022] [Indexed: 11/11/2022] Open
Abstract
The clinical presentation of MIS-C overlaps with other infectious/non-infectious diseases such as acute COVID-19, Kawasaki disease, acute dengue, enteric fever, and systemic lupus erythematosus. We examined the ex-vivo cellular parameters with the aim of distinguishing MIS-C from other syndromes with overlapping clinical presentations. MIS-C children differed from children with non-MIS-C conditions by having increased numbers of naïve CD8+ T cells, naïve, immature and atypical memory B cells and diminished numbers of transitional memory, stem cell memory, central and effector memory CD4+ and CD8+ T cells, classical, activated memory B and plasma cells and monocyte (intermediate and non-classical) and dendritic cell (plasmacytoid and myeloid) subsets. All of the above alterations were significantly reversed at 6-9 months post-recovery in MIS-C. Thus, MIS-C is characterized by a distinct cellular signature that distinguishes it from other syndromes with overlapping clinical presentations. Trial Registration: ClinicalTrials.gov clinicaltrial.gov. No: NCT04844242.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- National Institutes of Health-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
- * E-mail: (AR); (NPV)
| | - Pavan Kumar Nathella
- ICMR-National Institute for Research in Tuberculosis, Chennai, India
- * E-mail: (AR); (NPV)
| | | | | | - Srinithi Kannan
- Institute of Child Health and Hospital for Children, Chennai, India
| | - Arul Nancy Pandiarajan
- National Institutes of Health-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Rachel Mariam Renji
- National Institutes of Health-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | | | | | | | | | | | | | | | | | - Syed Hissar
- ICMR-National Institute for Research in Tuberculosis, Chennai, India
| | | | | | - Thomas B. Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Subash Babu
- National Institutes of Health-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
41
|
Kumar NP, Banurekha VV, Kumar CPG, Nancy A, Padmapriyadarsini C, Shankar S, Hanna LE, Murhekar M, Devi KRU, Babu S. Inactivated COVID-19 vaccines: durability of Covaxin/BBV152 induced immunity against variants of concern. J Travel Med 2022; 29:6651027. [PMID: 35900009 PMCID: PMC9384591 DOI: 10.1093/jtm/taac088] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Covaxin/BBV152 is one of the most widely used vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and one of the few vaccines used extensively in low- and middle-income countries (LMIC). METHODS We investigated the effect of Covaxin on the SARS-CoV-2 specific IgG and IgA and neutralizing antibody (NAb) levels at baseline (M0) and at Months 1 (M1), 2 (M2), 3 (M3), 4 (M4), 6 (M6) and 12 (M12) following vaccination in healthcare workers. In addition, we also examined the NAb levels against variant lineages of B.1.617.2 (Delta, India), B.1.617.2.1 (Delta Plus, India), B.1.351 (Beta, SA), B.1.1.7 (Alpha, UK) and B.1.1.529 (Omicron). RESULTS Covaxin induces enhanced SARS-CoV-2 binding antibodies of IgG and IgA responses against both spike (S) and nucleocapsid (N) antigens at M1, M2, M3, M4, M6 and M12 in comparison with M0. Our data also reveal that NAb levels against the ancestral strain (Wuhan, wild type) are elevated and sustained at M1, M2, M3, M4, M6 and M12 in comparison with M0 and against variant lineages of B.1.617.2 (Delta, India), B.1.617.2.1 (Delta Plus, India), B.1.351 (Beta, SA) and B.1.1.7 (Alpha, UK) are elevated at M3, M6 and M12 in comparison with M0. However, NAb levels against B.1.1.529 (Omicron) was consistently below the limit of detection except at M12. CONCLUSION Thus, Covaxin induces an enhanced humoral immune response, with persistence till at least 12 months post-vaccination against most SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Nathella Pavan Kumar
- Department of Immunology, ICMR-National Institute for Research in Tuberculosis, Chennai 600031, India
| | - V V Banurekha
- Department of Clinical Research, ICMR-National Institute for Research in Tuberculosis, Chennai 600031, India
| | - C P Girish Kumar
- Laboratory Division, ICMR-National Institute of Epidemiology, Chennai 600077, India
| | - Arul Nancy
- International Centre for Excellence in Research, ICMR-National Institute for Research in Tuberculosis, Chennai 600031, India
| | | | - Sakila Shankar
- Department of Clinical Research, ICMR-National Institute for Research in Tuberculosis, Chennai 600031, India
| | - Luke Elizabeth Hanna
- Department of Virology and Biotechnology, ICMR-National Institute for Research in Tuberculosis, Chennai 600031, India
| | - Manoj Murhekar
- Epidemiology and Biostatistics Division, ICMR-National Institute of Epidemiology, Chennai 600077, India
| | - K R Uma Devi
- Department of Immunology, ICMR-National Institute for Research in Tuberculosis, Chennai 600031, India
| | - Subash Babu
- International Centre for Excellence in Research, ICMR-National Institute for Research in Tuberculosis, Chennai 600031, India
| |
Collapse
|
42
|
Collins E, Galipeau Y, Arnold C, Bosveld C, Heiskanen A, Keeshan A, Nakka K, Shir-Mohammadi K, St-Denis-Bissonnette F, Tamblyn L, Vranjkovic A, Wood LC, Booth R, Buchan CA, Crawley AM, Little J, McGuinty M, Saginur R, Langlois MA, Cooper CL. Cohort profile: Stop the Spread Ottawa (SSO) -a community-based prospective cohort study on antibody responses, antibody neutralisation efficiency and cellular immunity to SARS-CoV-2 infection and vaccination. BMJ Open 2022; 12:e062187. [PMID: 36691221 PMCID: PMC9461086 DOI: 10.1136/bmjopen-2022-062187] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/16/2022] [Indexed: 01/27/2023] Open
Abstract
PURPOSE To investigate the robustness and longevity of SARS-CoV-2 immune responses conferred by natural infection and vaccination among priority populations such as immunocompromised individuals and people with post-acute sequelae of COVID-19 in a prospective cohort study (Stop the Spread Ottawa-SSO) in adults living in the Ottawa region. In this paper, we describe the study design, ongoing data collection and baseline characteristics of participants. PARTICIPANTS Since October 2020, participants who tested positive for COVID-19 (convalescents) or at high risk of exposure to the virus (under surveillance) have provided monthly blood and saliva samples over a 10-month period. As of 2 November 2021, 1026 adults had completed the baseline survey and 976 had attended baseline bloodwork. 300 participants will continue to provide bimonthly blood samples for 24 additional months (ie, total follow-up of 34 months). FINDINGS TO DATE The median age of the baseline sample was 44 (IQR 23, range: 18-79) and just over two-thirds (n=688; 67.1%) were female. 255 participants (24.9%) had a history of COVID-19 infection confirmed by PCR and/or serology. Over 600 participants (60.0%) work in high-risk occupations (eg, healthcare, teaching and transportation). 108 participants (10.5%) reported immunocompromising conditions or treatments at baseline (eg, cancer, HIV, other immune deficiency, and/or use of immunosuppressants). FUTURE PLANS SSO continues to yield rich research potential, given the collection of pre-vaccine baseline data and samples from the majority of participants, recruitment of diverse subgroups of interest, and a high level of participant retention and compliance with monthly sampling. The 24-month study extension will maximise opportunities to track SARS-CoV-2 immunity and vaccine efficacy, detect and characterise emerging variants, and compare subgroup humoral and cellular response robustness and persistence.
Collapse
Affiliation(s)
- Erin Collins
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Yannick Galipeau
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Corey Arnold
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Cameron Bosveld
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Aliisa Heiskanen
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Alexa Keeshan
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Kiran Nakka
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Khatereh Shir-Mohammadi
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Laura Tamblyn
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Agatha Vranjkovic
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Leah C Wood
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Ronald Booth
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Immunology Section, Eastern Ontario Regional Laboratory Association (EORLA), Ottawa, Ontario, Canada
| | - C Arianne Buchan
- Division of Infectious Diseases, Department of Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Angela M Crawley
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Coronavirus Variants Rapid Response Network (CoVaRR-Net), Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, Ottawa, Ontario, Canada
| | - Julian Little
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Coronavirus Variants Rapid Response Network (CoVaRR-Net), Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- The Knowledge Synthesis and Application Unit (KSAU), University of Ottawa, Ottawa, Ontario, Canada
| | - Michaeline McGuinty
- Division of Infectious Diseases, Department of Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Raphael Saginur
- Division of Infectious Diseases, Department of Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Health Science Network Research Ethics Board (OHSN-REB), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Coronavirus Variants Rapid Response Network (CoVaRR-Net), Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, Ottawa, Ontario, Canada
| | - Curtis L Cooper
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Division of Infectious Diseases, Department of Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Coronavirus Variants Rapid Response Network (CoVaRR-Net), Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
43
|
Olafsdottir TA, Bjarnadottir K, Norddahl GL, Halldorsson GH, Melsted P, Gunnarsdottir K, Ivarsdottir E, Olafsdottir T, Arnthorsson AO, Theodors F, Eythorsson E, Helgason D, Eggertsson HP, Masson G, Bjarnadottir S, Saevarsdottir S, Runolfsdottir HL, Olafsson I, Saemundsdottir J, Sigurdsson MI, Ingvarsson RF, Palsson R, Thorgeirsson G, Halldorsson BV, Holm H, Kristjansson M, Sulem P, Thorsteinsdottir U, Jonsdottir I, Gudbjartsson DF, Stefansson K. HLA alleles, disease severity, and age associate with T-cell responses following infection with SARS-CoV-2. Commun Biol 2022; 5:914. [PMID: 36068292 PMCID: PMC9446630 DOI: 10.1038/s42003-022-03893-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 08/25/2022] [Indexed: 12/12/2022] Open
Abstract
Memory T-cell responses following SARS-CoV-2 infection have been extensively investigated but many studies have been small with a limited range of disease severity. Here we analyze SARS-CoV-2 reactive T-cell responses in 768 convalescent SARS-CoV-2-infected (cases) and 500 uninfected (controls) Icelanders. The T-cell responses are stable three to eight months after SARS-CoV-2 infection, irrespective of disease severity and even those with the mildest symptoms induce broad and persistent T-cell responses. Robust CD4+ T-cell responses are detected against all measured proteins (M, N, S and S1) while the N protein induces strongest CD8+ T-cell responses. CD4+ T-cell responses correlate with disease severity, humoral responses and age, whereas CD8+ T-cell responses correlate with age and functional antibodies. Further, CD8+ T-cell responses associate with several class I HLA alleles. Our results, provide new insight into HLA restriction of CD8+ T-cell immunity and other factors contributing to heterogeneity of T-cell responses following SARS-CoV-2 infection. A study of 768 convalescent SARS CoV-2-infected and 500 uninfected Icelanders reveals broad and stable T-cell responses 3-8 months from infection. HLA alleles, disease severity, and age contribute to the heterogeneity of cellular immunity.
Collapse
Affiliation(s)
| | | | | | | | - Pall Melsted
- deCODE genetics/Amgen Inc., Reykjavik, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | | | | | - Elias Eythorsson
- Internal Medicine and Emergency Services, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | - Dadi Helgason
- Internal Medicine and Emergency Services, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | | | | | - Sólveig Bjarnadottir
- Internal Medicine and Emergency Services, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Saedis Saevarsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Department of Medicine, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Hrafnhildur L Runolfsdottir
- Internal Medicine and Emergency Services, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | - Isleifur Olafsson
- Clinical Laboratory Services, Diagnostics and Blood Bank, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | | | - Martin I Sigurdsson
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Perioperative Services, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | - Ragnar F Ingvarsson
- Internal Medicine and Emergency Services, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | - Runolfur Palsson
- Internal Medicine and Emergency Services, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Gudmundur Thorgeirsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Bjarni V Halldorsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland.,School of Science and Engineering, Reykjavik University, Reykjavík, Iceland
| | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | - Mar Kristjansson
- Internal Medicine and Emergency Services, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | | | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Ingileif Jonsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland. .,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
44
|
Lazarus R, Taucher C, Brown C, Čorbic Ramljak I, Danon L, Dubischar K, Duncan CJA, Eder-Lingelbach S, Faust SN, Green C, Gokani K, Hochreiter R, Wright JK, Kwon D, Middleditch A, Munro APS, Naker K, Penciu F, Price D, Querton B, Riaz T, Ross-Russell A, Sanchez-Gonzalez A, Wardle H, Warren S, Finn A. Safety and immunogenicity of the inactivated whole-virus adjuvanted COVID-19 vaccine VLA2001: A randomized, dose escalation, double-blind phase 1/2 clinical trial in healthy adults. J Infect 2022; 85:306-317. [PMID: 35718205 PMCID: PMC9212764 DOI: 10.1016/j.jinf.2022.06.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 06/11/2022] [Indexed: 01/02/2023]
Abstract
OBJECTIVES We aimed to evaluate the safety and optimal dose of a novel inactivated whole-virus adjuvanted vaccine against SARS-CoV-2: VLA2001. METHODS We conducted an open-label, dose-escalation study followed by a double-blind randomized trial using low, medium and high doses of VLA2001 (1:1:1). The primary safety outcome was the frequency and severity of solicited local and systemic reactions within 7 days after vaccination. The primary immunogenicity outcome was the geometric mean titre (GMT) of neutralizing antibodies against SARS-CoV-2 two weeks after the second vaccination. The study is registered as NCT04671017. RESULTS Between December 16, 2020, and June 3, 2021, 153 healthy adults aged 18-55 years were recruited in the UK. Overall, 81.7% of the participants reported a solicited AE, with injection site tenderness (58.2%) and headache (46.4%) being the most frequent. Only 2 participants reported a severe solicited event. Up to day 106, 131 (85.6%) participants had reported any AE. All observed incidents were transient and non-life threatening in nature. Immunogenicity measured at 2 weeks after completion of the two-dose priming schedule, showed significantly higher GMTs of SARS-CoV-2 neutralizing antibody titres in the highest dose group (GMT 545.6; 95% CI: 428.1, 695.4) which were similar to a panel of convalescent sera (GMT 526.9; 95% CI: 336.5, 825.1). Seroconversion rates of neutralizing antibodies were also significantly higher in the high-dose group (>90%) compared to the other dose groups. In the high dose group, antigen-specific IFN-γ expressing T-cells reactive against the S, M and N proteins were observed in 76, 36 and 49%, respectively. CONCLUSIONS VLA2001 was well tolerated in all tested dose groups, and no safety signal of concern was identified. The highest dose group showed statistically significantly stronger immunogenicity with similar tolerability and safety, and was selected for phase 3 clinical development.
Collapse
Affiliation(s)
- Rajeka Lazarus
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Christian Taucher
- Valneva Austria GmbH, Campus Vienna Biocenter 3, Vienna 1030, Austria.
| | - Claire Brown
- NIHR/Wellcome Trust Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Leon Danon
- Department of Engineering Mathematics, University of Bristol, Bristol, UK
| | - Katrin Dubischar
- Valneva Austria GmbH, Campus Vienna Biocenter 3, Vienna 1030, Austria
| | - Christopher J A Duncan
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle, UK
| | | | - Saul N Faust
- NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Christopher Green
- NIHR/Wellcome Trust Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Karishma Gokani
- NIHR/Wellcome Trust Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Romana Hochreiter
- Valneva Austria GmbH, Campus Vienna Biocenter 3, Vienna 1030, Austria
| | | | - Dowan Kwon
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | | | - Alasdair P S Munro
- NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Kush Naker
- NIHR/Wellcome Trust Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Florentina Penciu
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - David Price
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle, UK
| | - Benedicte Querton
- Valneva Austria GmbH, Campus Vienna Biocenter 3, Vienna 1030, Austria
| | - Tawassal Riaz
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Amy Ross-Russell
- NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Amada Sanchez-Gonzalez
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle, UK
| | - Hayley Wardle
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle, UK
| | - Sarah Warren
- NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Adam Finn
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK; Schools of Population Health Sciences and Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | | |
Collapse
|
45
|
Isnaini N, Mardian Y, Lokida D, Budiono F, Butar-Butar DP, Arlinda D, Salim G, Kosasih H, Wulan WN, Perodin J, Neal A, Lane HC, Karyana M. Mild reinfection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Delta variant: First case report from Indonesia. Front Med (Lausanne) 2022; 9:906469. [PMID: 35935779 PMCID: PMC9355687 DOI: 10.3389/fmed.2022.906469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/27/2022] [Indexed: 12/21/2022] Open
Abstract
Background Reinfection with SARS-CoV-2 has been well documented, yet little is known about the degree of protection a previous infection provides against reinfection, especially against Variants of Concern (VOC). Case presentation Here we describe a case of an unvaccinated 49-year-old man who experienced two sequential SARS-CoV-2 infections with two different variants, as evidenced by genomic sequencing. The first episode was caused by the Pango lineage B.1.466.2 and resulted in severe COVID-19 with 5 days in an intensive care unit (ICU). The second episode occurred approximately 6 months later, during the Delta surge in Indonesia. Genomic analysis showed that the second infection was caused by the Delta variant (Pango lineage B.1.617.2) and resulted in mild disease that did not require hospitalization. No SARS-CoV-2 nucleic acid was detected between the two episodes, but both binding and neutralizing antibodies to SARS-CoV-2 were detected prior to the reinfection, with the second infection leading to an increase in the levels of antibody. Conclusion We confirmed that the patient experienced a reinfection instead of persistent viral shedding from the first infection based on epidemiological, clinical, serological, and genomic analyses. Our case supports the hypothesis that SARS-CoV-2 reinfection may occur once antibody titers decrease or following the emergence of a new variant. The milder presentation in the patient’s second infection deserves further investigation to provide a clear picture of the role of post-infection immunity in altering the course of subsequent disease.
Collapse
Affiliation(s)
| | - Yan Mardian
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | - Dewi Lokida
- Tangerang District Hospital, Tangerang, Indonesia.,Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | | | - Deni P Butar-Butar
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | - Dona Arlinda
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia.,National Institute of Health Research and Development, Ministry of Health, Jakarta, Indonesia
| | - Gustiani Salim
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | - Herman Kosasih
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | - Wahyu Nawang Wulan
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | | | - Aaron Neal
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - H Clifford Lane
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Muhammad Karyana
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia.,National Institute of Health Research and Development, Ministry of Health, Jakarta, Indonesia
| |
Collapse
|
46
|
Pozdnyakova V, Weber B, Cheng S, Ebinger JE. Review of Immunologic Manifestations of COVID-19 Infection and Vaccination. Cardiol Clin 2022; 40:301-308. [PMID: 35851453 PMCID: PMC8960179 DOI: 10.1016/j.ccl.2022.03.006] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We herein summarize currently available and clinically relevant information regarding the human immune responses to SARS-CoV-2 infection and vaccination, in relation to COVID-19 outcomes with a focus on acute respiratory distress syndrome (ARDS) and myocarditis.
Collapse
Affiliation(s)
- Valeriya Pozdnyakova
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, D4005, Los Angeles, CA 90048, USA
| | - Brittany Weber
- Carl J. and Ruth Shapiro Cardiovascular Center, Brigham and Women's Hospital, 70 Francis Street, Boston, MA 02115, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South Vicente Boulevard, Suite A3100, Los Angeles, CA 90048, USA
| | - Joseph E Ebinger
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South Vicente Boulevard, Suite A3100, Los Angeles, CA 90048, USA.
| |
Collapse
|
47
|
Rebholz H, Braun RJ, Saha T, Harzer O, Schneider M, Ladage D. Longitudinal monitoring of SARS-CoV-2 spike protein-specific antibody responses in Lower Austria. PLoS One 2022; 17:e0271382. [PMID: 35895668 PMCID: PMC9328535 DOI: 10.1371/journal.pone.0271382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
The Lower Austrian Wachau region was an early COVID-19 hotspot of infection. As previously reported, in June 2020, after the first peak of infections, we determined that 8.5% and 9.0% of the participants in Weißenkirchen and surrounding communities in the Wachau region were positive for immunoglobulin G (IgG) and immunoglobulin A (IgA) antibodies against the receptor-binding domain of the spike protein of SARS-CoV-2, respectively. Here, we present novel data obtained eight months later (February 2021) from Weißenkirchen, after the second peak of infection, with 25.0% (138/552) and 23.6% (130/552) of participants that are positive for IgG and IgA, respectively. In participants with previous IgG/IgA positivity (June 2020), we observed a 24% reduction in IgG levels, whereas the IgA levels remained stable in February 2021. This subgroup was further analyzed for SARS-CoV-2 induced T cell activities. Although 76% (34/45) and 76% (34/45) of IgG positive and IgA positive participants, respectively, showed specific T cell activities (upon exposure to SARS-CoV-2 spike protein-derived peptides), those were not significantly correlated with the levels of IgG or IgA. Thus, the analyses of antibodies cannot surrogate the measurement of T cell activities. For a comprehensive view on SARS-CoV-2-triggered immune responses, the measurement of different classes of antibodies should be complemented with the determination of T cell activities.
Collapse
Affiliation(s)
- Heike Rebholz
- Research Division for Neurodegenerative Diseases, Center for Biosciences, Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR S1266, INSERM, Université de Paris, Paris, France
- GHU Psychiatrie et Neurosciences, Paris, France
| | - Ralf J. Braun
- Research Division for Neurodegenerative Diseases, Center for Biosciences, Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
- * E-mail: (RJB); (DL)
| | - Titas Saha
- Department of Scientific Coordination and Management, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
| | - Oliver Harzer
- Center for Biosciences, Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
- Bioscientia, Institute of Medical Diagnostics, Ingelheim, Germany
| | - Miriam Schneider
- Department of Scientific Coordination and Management, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
| | - Dennis Ladage
- Internal Medicine, Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
- Heart Center, University of Cologne, Cologne, Germany
- Department of Pneumology, Maria Hilf Hospital, Mönchengladbach, Germany
- * E-mail: (RJB); (DL)
| |
Collapse
|
48
|
Dhanya CR, Shailaja A, Mary AS, Kandiyil SP, Savithri A, Lathakumari VS, Veettil JT, Vandanamthadathil JJ, Madhavan M. RNA Viruses, Pregnancy and Vaccination: Emerging Lessons from COVID-19 and Ebola Virus Disease. Pathogens 2022; 11:800. [PMID: 35890044 PMCID: PMC9322689 DOI: 10.3390/pathogens11070800] [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: 05/26/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 02/01/2023] Open
Abstract
Pathogenic viruses with an RNA genome represent a challenge for global human health since they have the tremendous potential to develop into devastating pandemics/epidemics. The management of the recent COVID-19 pandemic was possible to a certain extent only because of the strong foundations laid by the research on previous viral outbreaks, especially Ebola Virus Disease (EVD). A clear understanding of the mechanisms of the host immune response generated upon viral infections is a prime requisite for the development of new therapeutic strategies. Hence, we present here a comparative study of alterations in immune response upon SARS-CoV-2 and Ebola virus infections that illustrate many common features. Vaccination and pregnancy are two important aspects that need to be studied from an immunological perspective. So, we summarize the outcomes and immune responses in vaccinated and pregnant individuals in the context of COVID-19 and EVD. Considering the significance of immunomodulatory approaches in combating both these diseases, we have also presented the state of the art of such therapeutics and prophylactics. Currently, several vaccines against these viruses have been approved or are under clinical trials in various parts of the world. Therefore, we also recapitulate the latest developments in these which would inspire researchers to look for possibilities of developing vaccines against many other RNA viruses. We hope that the similar aspects in COVID-19 and EVD open up new avenues for the development of pan-viral therapies.
Collapse
Affiliation(s)
| | - Aswathy Shailaja
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA;
| | - Aarcha Shanmugha Mary
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur 610105, India;
| | | | - Ambili Savithri
- Department of Biochemistry, Sree Narayana College, Kollam 691001, India;
| | | | | | | | - Maya Madhavan
- Department of Biochemistry, Government College for Women, Thiruvananthapuram 695014, India
| |
Collapse
|
49
|
Wang Y, Tian Q, Ye L. The Differentiation and Maintenance of SARS-CoV-2-Specific Follicular Helper T Cells. Front Cell Infect Microbiol 2022; 12:953022. [PMID: 35909969 PMCID: PMC9329515 DOI: 10.3389/fcimb.2022.953022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/20/2022] [Indexed: 12/24/2022] Open
Abstract
Upon acute viral infection, virus-specific CD4+ T cells differentiate into either TH1 cells or follicular helper T (TFH) cells. The molecular pathways governing such bimodal cell fate commitment remain elusive. Additionally, effector virus-specific TFH cells further differentiate into corresponding memory population, which confer long-term protection against re-infection of same viruses by providing immediate help to virus-specific memory B cells. Currently, the molecular mechanisms underlying the long-term maintenance of memory TFH cells are largely unknown. In this review, we discuss current understanding of early differentiation of virus-specific effector TFH cells and long-term maintenance of virus-specific memory TFH cells in mouse models of viral infection and patients of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection.
Collapse
Affiliation(s)
- Yifei Wang
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Qin Tian
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Institute of Immunology, The People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| | - Lilin Ye
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Institute of Immunology, The People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
- *Correspondence: Lilin Ye,
| |
Collapse
|
50
|
Howard FHN, Kwan A, Winder N, Mughal A, Collado-Rojas C, Muthana M. Understanding Immune Responses to Viruses-Do Underlying Th1/Th2 Cell Biases Predict Outcome? Viruses 2022; 14:1493. [PMID: 35891472 PMCID: PMC9324514 DOI: 10.3390/v14071493] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 12/15/2022] Open
Abstract
Emerging and re-emerging viral diseases have increased in number and geographical extent during the last decades. Examples include the current COVID-19 pandemic and the recent epidemics of the Chikungunya, Ebola, and Zika viruses. Immune responses to viruses have been well-characterised within the innate and adaptive immunity pathways with the outcome following viral infection predominantly attributed to properties of the virus and circumstances of the infection. Perhaps the belief that the immune system is often considered as a reactive component of host defence, springing into action when a threat is detected, has contributed to a poorer understanding of the inherent differences in an individual's immune system in the absence of any pathology. In this review, we focus on how these host factors (age, ethnicity, underlying pathologies) may skew the T helper cell response, thereby influencing the outcome following viral infection but also whether we can use these inherent biases to predict patients at risk of a deviant response and apply strategies to avoid or overcome them.
Collapse
Affiliation(s)
- Faith H. N. Howard
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK; (A.K.); (N.W.); (A.M.); (C.C.-R.); (M.M.)
| | | | | | | | | | | |
Collapse
|