1
|
Zhao F, Luo Y, Li B, Fan DD, Xiang Z, Li J, Zhong N, Chen R. Changed profiles of SARS-CoV-2 specific memory T cells in asthmatics with different blood eosinophil counts. Pulmonology 2025; 31:2424642. [PMID: 39883511 DOI: 10.1080/25310429.2024.2424642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025] Open
Affiliation(s)
- Fengming Zhao
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University. Guangzhou, Guangdong, P.R.China
| | - Yiting Luo
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University. Guangzhou, Guangdong, P.R.China
| | - Bizhou Li
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University. Guangzhou, Guangdong, P.R.China
| | - Dengxia Denise Fan
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ziyuan Xiang
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University. Guangzhou, Guangdong, P.R.China
| | - Jing Li
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University. Guangzhou, Guangdong, P.R.China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University. Guangzhou, Guangdong, P.R.China
- Guangzhou National Lab, Guangzhou, P.R.China
| | - Ruchong Chen
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University. Guangzhou, Guangdong, P.R.China
- Guangzhou National Lab, Guangzhou, P.R.China
| |
Collapse
|
2
|
Haltaufderhyde K, Gutiérrez AH, McAllister M, Boyle CM, Moise L, Martin W, De Groot AS. T-cell responses to highly conserved SARS-CoV-2 epitopes in Hispanic Americans receiving an mRNA COVID-19 vaccine. Hum Vaccin Immunother 2025; 21:2501844. [PMID: 40407705 PMCID: PMC12118426 DOI: 10.1080/21645515.2025.2501844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/02/2025] [Accepted: 05/01/2025] [Indexed: 06/01/2025] Open
Abstract
This study reports the pre-clinical evaluation of peptides from EPV-CoV-19, a T cell epitope-based SARS-CoV-2 vaccine candidate, following spike-mRNA vaccination of a predominantly Hispanic American cohort. EPV-COV-19 peptides' potential to boost T cell responses to spike protein vaccines was evaluated, confirming previously observed memory recall responses in donors with prior immunity to COVID-19. The vaccinated subjects' averaged immune responses to the 15-peptide EPV-CoV-19 pool achieved 85% of the observed response to a spike protein peptide array containing a 7-fold greater epitope content, suggesting that the EPV-CoV-19 peptides have a higher relative concentration of T cell epitope content per-peptide. Ten of the 15 peptides contained spike epitopes conserved in the majority of variants of concern (VOC) evaluated over the 2020-2024 period. While commercial vaccines exhibited gradual loss of T cell epitope conservation with VOC over time, the EPV-CoV-19 epitope-peptides maintained conservation until the XBB variant emerged. The addition of one new peptide to the vaccine design reestablished broad T cell epitope coverage. These findings underscore the importance of identifying highly conserved T cell epitopes for vaccine designs that target rapidly-mutating strains of emergent pathogens, while also documenting broad memory T cell response to the vaccine in a predominantly Hispanic American cohort.
Collapse
|
3
|
Alrubayyi A, Huang H, Gaiha GD. Severe Acute Respiratory Syndrome Coronavirus 2 Immunology and Coronavirus Disease 2019 Clinical Outcomes. Infect Dis Clin North Am 2025; 39:221-232. [PMID: 40089444 DOI: 10.1016/j.idc.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
The humoral and cellular immune response are the key players in preventing viral infection and limiting disease severity, particular in the context of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and coronavirus disease 2019. In this review, we discuss how immune responses generated by prior infection and vaccination influence the outcomes of SARS-CoV-2 infection. We aim to provide an overview of the role of humoral and cellular immunity, with a particular focus on CD8+ T cell responses, to delineate how different immune compartments contribute to the control of infection and modulation of disease outcomes.
Collapse
Affiliation(s)
| | - Hsinyen Huang
- Ragon Institute of MGB, MIT and Harvard, 600 Main Street, Cambridge, MA 02139
| | - Gaurav D Gaiha
- Ragon Institute of MGB, MIT and Harvard, 600 Main Street, Cambridge, MA 02139; Division of Gastroenterology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02115.
| |
Collapse
|
4
|
Lu HH, Dos Santos Alves RP, Li QH, Eder L, Timis J, Madany H, Chuensirikulchai K, Varghese KV, Singh A, Le Tran L, Street A, Elong Ngono A, Croft M, Shresta S. Enhanced durability of a Zika virus self-amplifying RNA vaccine through combinatorial OX40 and 4-1BB agonism. JCI Insight 2025; 10:e187405. [PMID: 40178907 DOI: 10.1172/jci.insight.187405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/28/2025] [Indexed: 04/05/2025] Open
Abstract
The SARS-CoV-2 pandemic highlighted the potential of mRNA vaccines in rapidly responding to emerging pathogens. However, immunity induced by conventional mRNA vaccines wanes quickly, requiring frequent boosters. Self-amplifying RNA (saRNA) vaccines, which extend antigen expression via self-replication, offer a promising strategy to induce more durable immune responses. In this study, we developed an saRNA vaccine encoding Zika virus (ZIKV) membrane and envelope proteins and evaluated its efficacy in mice. A single vaccination elicited strong humoral and cellular immune responses and reduced viral loads but only for 28 days. By day 84, antibody titers and T cell responses had significantly declined, resulting in reduced efficacy. To address this, we evaluated agonist antibodies targeting the T cell costimulatory molecules OX40 and 4-1BB. Coadministration of agonist antibodies enhanced CD8+ T cell responses to vaccination, resulting in sustained immunity and reduced viral loads at day 84. Depletion and passive transfer studies verified that long-term antiviral immunity was primarily CD8+ T cell dependent, with minimal contributions from antibody responses. These findings suggest that agonists targeting members of the tumor necrosis receptor superfamily, such as OX40 and 4-1BB, might enhance the durability of saRNA vaccine-induced protection, addressing a key limitation of current mRNA vaccine platforms.
Collapse
Affiliation(s)
- Hsueh-Han Lu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
| | | | - Qin Hui Li
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
| | - Luke Eder
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Julia Timis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Henry Madany
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | | | - Krithik V Varghese
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Aditi Singh
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Linda Le Tran
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Audrey Street
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Annie Elong Ngono
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
5
|
Jagne YJ, Jobe D, Darboe A, Danso M, Barratt N, Gomez M, Wenlock R, Jarju S, Sylva EL, Touray AF, Toure F, Kumado M, Saso A, Zafred D, Nicklin M, Sayers J, Hornsby H, Lindsey B, Sesay AK, Temperton N, Kucharski A, Hodgson D, de Silva T, Kampmann B. Compartmentalised mucosal and blood immunity to SARS-CoV-2 is associated with high seroprevalence before the Delta wave in Africa. COMMUNICATIONS MEDICINE 2025; 5:178. [PMID: 40379979 DOI: 10.1038/s43856-025-00902-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 05/09/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND The reported number of SARS-CoV-2 cases and deaths are lower in Africa compared to many high-income countries. However, in African cohorts, detailed characterisation of SARS-CoV-2 mucosal and T cell immunity are limited. We assessed the SARS-CoV-2-specific immune landscape in The Gambia during the presence of the pre-Delta variant in July 2021. METHODS A cross-sectional assessment of SARS-CoV-2 immunity in 349 unvaccinated individuals from 52 Gambian households was performed between March-June 2021. SARS-CoV-2 spike (S) and nucleocapsid (N) specific binding antibodies were measured by ELISA, variant-specific serum neutralizing-antibodies (NAb) by viral pseudotype assays and nasal fluid IgA by mesoscale discovery assay. SARS-CoV-2 T-cell responses were evaluated using ELISpot assay. RESULTS We show that adjusted anti-Spike antibody seroprevalence is 56.7% (95% confidence interval (CI) 49.0-64.0), with lower rates in children <5 years (26.2%, 13.9-43.8) and 5-17 years (46.4%, 36.2-56.7) compared to adults 18-49 years (78.4%, 68.8-85.8). Among spike-seropositive individuals, NAb titres are highest against Alpha variant (median IC50 110), with 27% showing pre-existing Delta variant titres >1:50. T-cell responses are higher in spike-seropositive individuals, although 34% of spike-seronegative individuals show responses to at least one antigen pool. We observe strong correlations within SARS-CoV-2 T-cell, mucosal IgA, and serum NAb responses. CONCLUSIONS High SARS-CoV-2 seroprevalence in The-Gambia induce mucosal and blood immunity, reducing Delta and Omicron impact. Children are relatively protected from infection. T-cell responses in seronegative individuals may indicate either pre-pandemic cross-reactivity or individuals with a T-cell dominated response to SARS-CoV-2 infection with absent or poor humoral responses.
Collapse
Affiliation(s)
- Ya Jankey Jagne
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia.
| | - Dawda Jobe
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Alansana Darboe
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Madikoi Danso
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Natalie Barratt
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
- The Florey Institute of Infection, The University of Sheffield, Sheffield, UK
| | - Marie Gomez
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Rhys Wenlock
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Sheikh Jarju
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Ellen Lena Sylva
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Aji Fatou Touray
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Fatoumata Toure
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Michelle Kumado
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Anja Saso
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Domen Zafred
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Martin Nicklin
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Jon Sayers
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Hailey Hornsby
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
- The Florey Institute of Infection, The University of Sheffield, Sheffield, UK
| | - Benjamin Lindsey
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
- The Florey Institute of Infection, The University of Sheffield, Sheffield, UK
| | - Abdul Karim Sesay
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Canterbury, UK
| | - Adam Kucharski
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | - David Hodgson
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | - Thushan de Silva
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
- The Florey Institute of Infection, The University of Sheffield, Sheffield, UK
| | - Beate Kampmann
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia.
- Charité Centre for Global Health; Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
6
|
Konopka EN, Edgerton AO, Kutzler MA. Nucleic acid vaccines: innovations, efficacy, and applications in at-risk populations. Front Immunol 2025; 16:1584876. [PMID: 40438110 PMCID: PMC12116436 DOI: 10.3389/fimmu.2025.1584876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/09/2025] [Indexed: 06/01/2025] Open
Abstract
For more than two centuries, the field of vaccine development has progressed through the adaptation of novel platforms in parallel with technological developments. Building off the advantages and shortcomings of first and second-generation vaccine platforms, the advent of third-generation nucleic acid vaccines has enabled new approaches to tackle emerging infectious diseases, cancers, and pathogens where vaccines remain unavailable. Unlike traditional vaccine platforms, nucleic acid vaccines offer several new advantages, including their lower cost and rapid production, which was widely demonstrated during the COVID-19 pandemic. Beyond production, DNA and mRNA vaccines can elicit unique and targeted responses through specialized design and delivery approaches. Considering the growth of nucleic acid vaccine research over the past two decades, the evaluation of their efficacy in at-risk populations is paramount for refining and improving vaccine design. Importantly, the aging population represents a significant portion of individuals highly susceptible to infection and disease. This review seeks to outline the major impairments in vaccine-induced responses due to aging that may be targeted for improvement with design and delivery components encompassing mRNA and DNA vaccine formulations. Results of pre-clinical and clinical applications of these vaccines in aged animal models and humans will also be evaluated to outline current successes and limitations observed in these platforms.
Collapse
Affiliation(s)
- Emily N. Konopka
- Drexel University College of Medicine, Department of Microbiology and Immunology, Philadelphia, PA, United States
- Drexel University College of Medicine, Department of Medicine, Division of Infectious Diseases and HIV Medicine, Philadelphia, PA, United States
| | - Arden O. Edgerton
- Drexel University College of Medicine, Department of Microbiology and Immunology, Philadelphia, PA, United States
- Drexel University College of Medicine, Department of Medicine, Division of Infectious Diseases and HIV Medicine, Philadelphia, PA, United States
| | - Michele A. Kutzler
- Drexel University College of Medicine, Department of Microbiology and Immunology, Philadelphia, PA, United States
- Drexel University College of Medicine, Department of Medicine, Division of Infectious Diseases and HIV Medicine, Philadelphia, PA, United States
| |
Collapse
|
7
|
Li X, Peng H, Wang Y, He S, Yang X, Chen J. The subsets of blood circulating T-cells associated with the development and prognosis of coinfection in patients with critical COVID-19. Front Immunol 2025; 16:1586302. [PMID: 40416960 PMCID: PMC12098604 DOI: 10.3389/fimmu.2025.1586302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/22/2025] [Indexed: 05/27/2025] Open
Abstract
Background A secondary bacterial infection, which has a high incidence in patients with critical coronavirus disease 2019 (COVID-19), has been proven to have an association with increased mortality. Adaptive immune responses have been detected in almost all COVID-19 cases. This study aimed to determine whether the levels of immune-inflammatory factors are associated with coinfection in patients with critical COVID-19. Methods Patients with a confirmed critical severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection were enrolled in this single-center cohort study. Clinical data and venous blood samples were collected on the day of hospital admission. All patients were divided into two groups according to the presence of bacterial coinfection or absence of bacterial coinfection, which were then divided into two groups (survived group and deceased group) based on the outcome of the disease during hospitalization. Results Patients with coinfection had a higher mortality rate (83.3% VS 50.0%, P<0.001) and longer hospital stays (25.15 VS 13.80d, P<0.001). We observed that patients who developed coinfection tended to have a significantly lower number of CD4+ T cells (121.19 VS 207.83cells/µL, P=0.001) and CD8+ T cells (79 VS 158cells/µL, P=0.006) and a higher proportion of CD4+CD8+ double-positive T (DPT) cells (3.66% VS 1.91%, P=0.011) on the day of hospital admission. The tests for inflammatory cytokines showed a higher level of IL-4 (0.99 VS 0.42pg/mL, P<0.001) and IL-6 (109.60 VS 63.59pg/mL, P=0.009) in coinfection group. And the multivariant analyses also revealed that CD4+ cell counts < 199.5cells/µL, CD8+ cell counts < 124.5cells/µL, IL4 > 0.535pg/mL, IL6 > 388.9pg/mL could be independent risk factors for coinfection. Moreover, in the coinfection group, we observed that the deceased patients had a lower level of total lymphocytes, T cells, and albumin. Conclusion Our study found that lymphocyte subsets and cytokines play an important role in predicting bacterial coinfection in patients with critical COVID-19. Lower levels of CD4+ and CD8+ cells and higher level of IL4 and IL6 in patients on the day of admission were significantly correlated with the development of coinfection the following days in the hospital.
Collapse
Affiliation(s)
- Xingming Li
- Department of Emergency, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, Chengdu, China
| | - Hongqiong Peng
- Department of Emergency, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, Chengdu, China
| | - Yunchuan Wang
- Department of Emergency, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, Chengdu, China
| | - Shiying He
- Department of Emergency, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, Chengdu, China
| | - Xueting Yang
- Department of Emergency, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, Chengdu, China
| | - Jiayue Chen
- Department of Pathology, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
8
|
Luo Q, Song Q, Li Y, Zong K, Liu T, He J, Mei G, Du H, Xia Z, Liu M, Song J, Gao C, Xia D, Xue G, Tian W, Qu Y, Kou Z, Dong Z, Han J. Reduced immune response to SARS-CoV-2 infection in the elderly after 6 months. Front Immunol 2025; 16:1596065. [PMID: 40416973 PMCID: PMC12098630 DOI: 10.3389/fimmu.2025.1596065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 04/11/2025] [Indexed: 05/27/2025] Open
Abstract
Objectives To evaluate the immune persistence and cross-immune response of elderly individuals after Omicron BA.5 infections. Method The neutralizing antibodies against WT, BA.5, XBB.1 and EG.5 strains were analyzed. The T/B-cell subsets' responses were tested through intracellular cytokine staining and flow cytometry. Results The neutralizing antibodies titers against WT and BA.5 strain, remaining high level for at least 6 months, were higher than that of both XBB.1 and EG.5 variants. The neutralizing antibodies of WT, BA.5, XBB.1, and EG.5 strains in the elderly were slightly lower than those in middle-age. The memory B cells decreased rapidly in the elderly, and Tfh, Th17 cells of the elderly continued to increase for only 3 months, while Tfh and Th17 cells increased in the middle-aged for over 6 months. For the elderly, after peptide stimulation, unswitched/switched memory B cells decreased, while double negative B cells displayed higher proliferation. The proportions of both naïve and Temra cells in CD4+ and CD8+ T cells declined, whereas those of Tcm and Tem cells elevated. In the meantime, both CD69+ and CD38+ T cells decreased, but the frequencies of PD-1+ and CTLA-4+ of CD4+ and CD8+ T cells showed an increasing trend. The proportions of PD-1+ and CTLA-4+ cells also increased in older people with long COVID symptoms at 3m post-infection. Conclusions Omicron BA.5 infection induced lower neutralizing antibodies against XBB.1 and EG.5 variant. The decrease of memory B cells, CD69+ and CD38+T cells, as well as the increase of PD-1+, CTLA-4+ of CD4+/CD8+T cells and double negative B cells, indicate that sustained immune responses against BA.5 infection may wane more rapidly in elderly populations.
Collapse
Affiliation(s)
- Qin Luo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qinqin Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Li
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Intelligent Monitoring, Early Warning, and Prevention and Control of Infectious Diseases, Shandong Institute of Preventive Medicine, Jinan, China
| | - Kexin Zong
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ti Liu
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Intelligent Monitoring, Early Warning, and Prevention and Control of Infectious Diseases, Shandong Institute of Preventive Medicine, Jinan, China
| | - Junming He
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Guoyong Mei
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Haijun Du
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhiqiang Xia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Mi Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Juan Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chen Gao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dong Xia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guangyu Xue
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wenyan Tian
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yinli Qu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zengqiang Kou
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Intelligent Monitoring, Early Warning, and Prevention and Control of Infectious Diseases, Shandong Institute of Preventive Medicine, Jinan, China
| | - Zhongjun Dong
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical Immunology, Anhui Medical University, Hefei, China
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, Beijing, China
| | - Jun Han
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
9
|
Ishmatov A. Age, gender, and race differences in nasal morphology: Linking air conditioning and filtration efficiency to disparities in air pollution health outcomes and COVID-19 mortality. CHEMOSPHERE 2025; 377:144358. [PMID: 40153988 DOI: 10.1016/j.chemosphere.2025.144358] [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: 12/24/2024] [Revised: 03/17/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
COVID-19 mortality disparities underscore the critical role of environmental factors, age, sex, and racial demographics. This study investigates how individual variations in nasal morphology - specifically its air conditioning (temperature and humidity regulation) and filtration functions - may influence respiratory health and contribute to differential COVID-19 outcomes. Analysis reveals significant differences in nasal structure and function across racial, sex, and age groups, demonstrating associations with disparities in respiratory vulnerability to environmental stressors such as air pollution, infectious aerosols, and climatic conditions. Specifically, wider nasal cavities (more common in certain populations), larger male nasal passages, and age-related changes like mucosal atrophy and increased endonasal volume impair air conditioning and filtration efficiency. These morphological variations influence the nose's protective capacity, which is critical for shielding the middle and lower airways from environmental exposures. Populations with inherently reduced nasal filtration and conditioning efficiency demonstrate higher vulnerability, aligning with U.S. mortality patterns for both COVID-19 and air pollution across demographic groups. This suggests a direct link between nasal anatomy and population-level health disparities. These findings provide novel insights into the role of nasal anatomy in mediating respiratory health disparities by modulating individual responses to environmental exposures, air pollution, and pathogens. They highlight the need to address critical gaps in understanding how airway characteristics influence susceptibility to environmental stressors and to develop targeted interventions aimed at reducing health disparities.
Collapse
Affiliation(s)
- Alexander Ishmatov
- Institute for Engineering and Environmental Safety, Togliatti State University, Belorusskaya St, 14, Togliatti, 445020, Russia.
| |
Collapse
|
10
|
Movsisyan M, Harutyunyan H, Movsisyan K, Kasparova I, Hakobyan A, Yenkoyan K. Age-related peculiarities of antibody-mediated humoral immune response following SARS-CoV-2 infection. Exp Gerontol 2025; 203:112735. [PMID: 40120835 DOI: 10.1016/j.exger.2025.112735] [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: 11/23/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Thousands of articles were published about the COVID-19 disease and hundreds about the immune response. But still little is known about the features of SARS-CoV-2-specific immunity in elderly. The aim of current research was to evaluate the age-related peculiarities of antibody mediated humoral immune response following SARS-CoV-2 infection. Our study presents an intriguing divergence from the classical concept of immunosenescence, where aging has been assumed to cause poor antibody responses, reduced or inefficient vaccination, and overall blunted immune responses in elderly people. Our findings were opposite to some of these expectations; participants aged over 60 expressed elevated titers of anti-SARS-CoV-2 antibodies in comparison to younger adults. Analyzing the data of relative neutralization and avidity of anti-SARS-Cov-2 (S) antibodies we propose that although older adults produce a higher quantity of antibodies, their functional efficiency appears relatively reduced exhibiting lower neutralizing capacity and binding strength per antibody compared to younger adults. We can assume that the immune system of the elderly may require a higher level of antibody production to obtain a comparable level of protection. Our findings highlight the intricate nature of immune responses in convalescent older adults. This has particular relevance to understanding immunity and vaccine responses in different age groups.
Collapse
Affiliation(s)
- M Movsisyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia; Department of Allergology and Clinical Immunology, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia
| | - H Harutyunyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia
| | - Kh Movsisyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia
| | - I Kasparova
- Department of Histology, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia
| | - A Hakobyan
- Department of Allergology and Clinical Immunology, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia
| | - K Yenkoyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia; Department of Biochemistry, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia.
| |
Collapse
|
11
|
Sedaghat-Rostami E, Carr BV, Yang L, Keep S, Lean FZX, Atkinson I, Fones A, Paudyal B, Kirk J, Vatzia E, Gubbins S, Bickerton E, Briggs E, Núñez A, McNee A, Moffat K, Freimanis G, Rollier C, Muir A, Richard AC, Angelopoulos N, Gerner W, Tchilian E. Porcine respiratory coronavirus as a model for acute respiratory disease: mechanisms of different infection outcomes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf066. [PMID: 40304579 DOI: 10.1093/jimmun/vkaf066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/11/2025] [Indexed: 05/02/2025]
Abstract
Porcine respiratory coronavirus (PRCV) is a naturally occurring pneumotropic coronavirus in the pig, providing a valuable large animal model to study acute respiratory disease. PRCV pathogenesis and the resulting immune response were investigated in pigs, the natural large animal host. We compared 2 strains, ISU-1 and 135, which induced differing levels of pathology in the respiratory tract to elucidate the mechanisms leading to mild or severe disease. The 135 strain induced greater pathology which was associated with higher viral load and stronger spike-specific antibody and T-cell responses. In contrast, the ISU-1 strain triggered mild pathology with a more balanced immune response and greater abundance of T regulatory cells. A higher frequency of putative T follicular helper cells was observed in animals infected with strain 135 at 11 days postinfection. Single-cell RNA-sequencing of bronchoalveolar lavage revealed differential gene expression in B and T cells between animals infected with 135 and ISU-1 at 1 day postinfection. These genes were associated with cell adhesion, migration, and immune regulation. Along with increased IL-6 and IL-12 production, these data indicate that heightened inflammatory responses to the 135 strain may contribute to pronounced pneumonia. Among bronchoalveolar lavage (BAL) immune cell populations, B cells and plasma cells exhibited the most gene expression divergence between pigs infected with different PRCV strains, highlighting their role in maintaining immune homeostasis in the respiratory tract. These findings indicate the potential of the PRCV model for studying coronavirus-induced respiratory disease and identifying mechanisms that determine infection outcomes.
Collapse
Affiliation(s)
- Ehsan Sedaghat-Rostami
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guilford, United Kingdom
| | | | - Liu Yang
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Sarah Keep
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Fabian Z X Lean
- Department of Pathology, Animal and Plant Health Agency, Addlestone, United Kingdom
| | - Isabella Atkinson
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Albert Fones
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Basudev Paudyal
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - James Kirk
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Eleni Vatzia
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Simon Gubbins
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Erica Bickerton
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Emily Briggs
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guilford, United Kingdom
| | - Alejandro Núñez
- Department of Pathology, Animal and Plant Health Agency, Addlestone, United Kingdom
| | - Adam McNee
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Katy Moffat
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Graham Freimanis
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Christine Rollier
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guilford, United Kingdom
| | - Andrew Muir
- Department of Pathology, Animal and Plant Health Agency, Addlestone, United Kingdom
| | | | - Nicos Angelopoulos
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Wilhelm Gerner
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Elma Tchilian
- Host response, The Pirbright Institute, Pirbright, Woking, United Kingdom
| |
Collapse
|
12
|
Zheng MZM, Burmas L, Tan HX, Trieu MC, Lee HJ, Rawlinson D, Haque A, Kent SJ, Wheatley AK, Juno JA. Deconvoluting TCR-dependent and -independent activation is vital for reliable Ag-specific CD4 + T cell characterization by AIM assay. SCIENCE ADVANCES 2025; 11:eadv3491. [PMID: 40279430 PMCID: PMC12024690 DOI: 10.1126/sciadv.adv3491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/20/2025] [Indexed: 04/27/2025]
Abstract
Activation-induced marker (AIM) assays identify antigen (Ag)-specific T cells, but recent studies revealed AIM+ T helper cell 17 (TH17)-like (CCR6+) and circulating T follicular helper cells (cTfh) were not associated with peptide/HLA tetramer staining. We show that CD39+ regulatory T cell (Treg)-like and CD26hi TH22-like cells undergo T cell receptor (TCR)-independent activation by cytokines during Ag stimulation, leading to nonspecific up-regulation of AIM readouts. Transcriptional analysis enabled discrimination of bona fide Ag-specific T cells from cytokine-activated Treg and TH22 cells. CXCR4 down-regulation emerged as a hallmark of clonotypic expansion and TCR-dependent activation in memory CD4+ T cells and cTfh. By tracking tetramer-binding cells upon Ag restimulation, we demonstrated that CXCR4-CD137+ cells provided a more accurate measure of Ag-specificity than standard AIM readouts. This modified assay excluded the predominantly CCR6+ cytokine-activated T cells that contributed to an average 12-fold overestimation of the Ag-specific population. Our findings provide an accurate approach to characterize genuine Ag-specific T cells.
Collapse
Affiliation(s)
- Ming Z. M. Zheng
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Lauren Burmas
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Mai-Chi Trieu
- Department of Clinical Science, Influenza Centre, University of Bergen and Haukeland University Hospital, Bergen, Norway
| | - Hyun Jae Lee
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Daniel Rawlinson
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
- Melbourne Integrative Genomics, School of Mathematics and Statistics, The University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Ashraful Haque
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria 3053, Australia
| | - Adam K. Wheatley
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Jennifer A. Juno
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| |
Collapse
|
13
|
Fan L, Qiu Z, Deng Q, Guo T, Rong L. Modeling SARS-CoV-2 Infection Dynamics: Insights into Viral Clearance and Immune Synergy. Bull Math Biol 2025; 87:67. [PMID: 40232610 DOI: 10.1007/s11538-025-01442-0] [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: 09/12/2024] [Accepted: 03/18/2025] [Indexed: 04/16/2025]
Abstract
Understanding the mechanisms of interaction between SARS-CoV-2 infection and the immune system is crucial for developing effective treatment strategies against COVID-19. In this paper, a mathematical model is formulated to investigate the interactions among SARS-CoV-2 infection, cellular immunity, and humoral immunity. Clinical data from eight asymptomatic or mild COVID-19 patients in Munich are used to fit the model, and the dynamics of natural killer (NK) cells, cytotoxic T lymphocytes (CTLs), B cells, and antibodies are further explored using the average of the best-fitting parameter values. Subsequently, the impact of NK cells, CTLs, B cells, and antibodies on SARS-CoV-2 infection is numerically investigated. The results indicate that (i) the synergy of NK cells, CTLs, and antibodies leads to a rapid decrease in the viral load during SARS-CoV-2 infection; (ii) antibodies play a crucial role compared to other immune mechanisms, and enhancing B cell stimulation may be more effective in clearing the virus from the lungs; (iii) in terms of cytotoxic effects, CTLs are stronger and more sustained than NK cells. Furthermore, the existence and local stability of the model's equilibria are fully classified, and complex dynamics of the model are further investigated using bifurcation theory, revealing multistability phenomena, including multiple attractors and periodic solutions. These findings suggest potential uncertainty and diversity in SARS-CoV-2 infection outcomes.
Collapse
Affiliation(s)
- Lele Fan
- School of Mathematics and Statistics, Nanjing University of Science and Technology, Nanjing, 210094, People's Republic of China
| | - Zhipeng Qiu
- School of Mathematics and Statistics, Nanjing University of Science and Technology, Nanjing, 210094, People's Republic of China.
| | - Qi Deng
- Laboratory for Industrial and Applied Mathematics, Department of Mathematics and Statistics, York University, Toronto, ON, M3J1P0, Canada
| | - Ting Guo
- Aliyun School of Big Data, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Libin Rong
- Department of Mathematics, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
14
|
Zhang J, Fang F, Zhang Y, Han X, Wang Y, Yin Q, Sun K, Zhou H, Qin H, Zhao D, Tai W, Zhang J, Zhang Z, Yang T, Wei Y, Zhang S, Li S, Li M, Zhao G. Humanized Major Histocompatibility Complex Transgenic Mouse Model Can Play a Potent Role in SARS-CoV-2 Human Leukocyte Antigen-Restricted T Cell Epitope Screening. Vaccines (Basel) 2025; 13:416. [PMID: 40333292 PMCID: PMC12031200 DOI: 10.3390/vaccines13040416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/25/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
Background: COVID-19, caused by SARS-CoV-2, poses a significant threat to human health. Vaccines designed for T-cell epitopes play an important role in eliminating the virus. However, T cell epitope screening often requires the use of a large number of peripheral blood mononuclear cells (PBMCs) from infected or convalescent patients, and if MHC humanized mice can be used for epitope screening, they will not have to wait for enough PBMCs to be available to screen for epitopes, thus buying time for epitope confirmation and vaccine design. Methods: In this study, we used SARS-CoV-2 BA.5 to infect HLA-A11/DR1, C57BL/6, hACE2 mice, and detected body weight changes, viral load, and pathological changes after infection. Fourteen days after the HLA-A11/DR1 and C57BL/6 mice were immunized against inactivated viruses, IgG antibodies were detected in mouse serum using ELISA, and IFN-γ produced by peptide stimulation of splenocytes was detected by ELISpot. Results: There is no obvious pathogenic phenotype of SARS-CoV-2 infection in HLA-A11/DR1 mice. Specific IgG antibodies were detected in serum after immunization of inactivated virus in both HLA-A11/DR1 and C57BL/6 mice, but specific IFN-γ was detected in splenocytes of HLA-A11/DR1 mice. Conclusions: Although HLA-A11/DR1 mice are unable to replicate the virus effectively in vivo, they are able to generate cellular immune responses after immunization inactivated viruses. Therefore, it can be used as a tool to substitute for human PBMCs in epitope screening, thus shortening the timeliness of T cell epitope screening and obtaining the immunogenicity information of new epitopes in a timely manner.
Collapse
Affiliation(s)
- Jiejie Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (J.Z.); (F.F.); (H.Z.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (Q.Y.); (K.S.); (T.Y.); (Y.W.); (S.Z.)
| | - Feimin Fang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (J.Z.); (F.F.); (H.Z.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (Q.Y.); (K.S.); (T.Y.); (Y.W.); (S.Z.)
| | - Yue Zhang
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China; (Y.Z.); (J.Z.); (Z.Z.)
| | - Xuelian Han
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (Q.Y.); (K.S.); (T.Y.); (Y.W.); (S.Z.)
| | - Yuan Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (Q.Y.); (K.S.); (T.Y.); (Y.W.); (S.Z.)
| | - Qi Yin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (Q.Y.); (K.S.); (T.Y.); (Y.W.); (S.Z.)
| | - Keyu Sun
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (Q.Y.); (K.S.); (T.Y.); (Y.W.); (S.Z.)
- Public Health School, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Haisheng Zhou
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (J.Z.); (F.F.); (H.Z.)
| | - Hanxiong Qin
- Changchun Institute of Biological Products Co., Ltd., Changchun 130012, China; (H.Q.); (D.Z.)
| | - Dongmei Zhao
- Changchun Institute of Biological Products Co., Ltd., Changchun 130012, China; (H.Q.); (D.Z.)
| | - Wanbo Tai
- Shenzhen Bay Laboratory, Institute of Infectious Diseases, Shenzhen 518132, China;
| | - Jun Zhang
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China; (Y.Z.); (J.Z.); (Z.Z.)
| | - Zhang Zhang
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China; (Y.Z.); (J.Z.); (Z.Z.)
| | - Tiantian Yang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (Q.Y.); (K.S.); (T.Y.); (Y.W.); (S.Z.)
- Public Health School, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Yuwei Wei
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (Q.Y.); (K.S.); (T.Y.); (Y.W.); (S.Z.)
| | - Shuai Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (Q.Y.); (K.S.); (T.Y.); (Y.W.); (S.Z.)
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Shuai Li
- Changchun Institute of Biological Products Co., Ltd., Changchun 130012, China; (H.Q.); (D.Z.)
| | - Min Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (Q.Y.); (K.S.); (T.Y.); (Y.W.); (S.Z.)
| | - Guangyu Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (J.Z.); (F.F.); (H.Z.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China; (Y.Z.); (J.Z.); (Z.Z.)
| |
Collapse
|
15
|
Stone S, Elsharkawy A, Burleson JD, Hauser M, Domi A, Kumari P, Nabi Z, Natekar JP, Porto M, Backstedt B, Newman M, Oruganti SR, Kumar M. Multi-Antigen Viral-Vectored Vaccine Protects Against SARS-CoV-2 and Variants in a Lethal hACE2 Transgenic Mouse Model. Vaccines (Basel) 2025; 13:411. [PMID: 40333327 PMCID: PMC12031414 DOI: 10.3390/vaccines13040411] [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: 02/05/2025] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025] Open
Abstract
Widespread and rapidly evolving SARS-CoV-2 posed an unprecedented challenge to vaccine developers. GeoVax has designed a multiantigen SARS-CoV-2 vaccine, designated GEO-CM02 based on a Modified Vaccinia Virus (MVA) vector that expresses spike (S), membrane (M), and envelope (E) antigens. This experimental vaccine was tested in the hACE2 transgenic mouse model to assess immunogenicity and efficacy. Administration of the vaccine in a two-dose regimen elicited high levels of neutralizing antibodies and provided complete protection, effectively reducing lung, olfactory bulb, and brain viral load and reducing lung inflammation following infection with original B.1 virus and the B.1.1.529 variant. In addition, GEO-CM02 conferred 80% protection against a lethal infection with the B.1.351 variant. GEO-CM02 vaccine efficacy studies also demonstrated a complete level of vaccine-induced protection with a single dose against the original B.1 virus and B.1.1.529 variant. GEO-CM02 effectively elicited functional T-cell responses in both prime and prime-boost groups. These data indicate that vaccination with the GEO-CM02 vaccine can induce immune responses that protect against severe disease induced by SARS-CoV-2 and its variants in a highly relevant pre-clinical model.
Collapse
Affiliation(s)
- Shannon Stone
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.S.); (A.E.); (Z.N.); (J.P.N.)
| | - Amany Elsharkawy
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.S.); (A.E.); (Z.N.); (J.P.N.)
| | - J. D. Burleson
- GeoVax, Inc., Atlanta, GA 30080, USA; (J.D.B.); (M.H.); (A.D.); (P.K.); (M.N.)
| | - Mary Hauser
- GeoVax, Inc., Atlanta, GA 30080, USA; (J.D.B.); (M.H.); (A.D.); (P.K.); (M.N.)
| | - Arban Domi
- GeoVax, Inc., Atlanta, GA 30080, USA; (J.D.B.); (M.H.); (A.D.); (P.K.); (M.N.)
| | - Pratima Kumari
- GeoVax, Inc., Atlanta, GA 30080, USA; (J.D.B.); (M.H.); (A.D.); (P.K.); (M.N.)
| | - Zainab Nabi
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.S.); (A.E.); (Z.N.); (J.P.N.)
| | - Janhavi P. Natekar
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.S.); (A.E.); (Z.N.); (J.P.N.)
| | - Maciel Porto
- BioQual, Inc., Rockville, MD 20850, USA; (M.P.); (B.B.)
| | | | - Mark Newman
- GeoVax, Inc., Atlanta, GA 30080, USA; (J.D.B.); (M.H.); (A.D.); (P.K.); (M.N.)
| | | | - Mukesh Kumar
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.S.); (A.E.); (Z.N.); (J.P.N.)
| |
Collapse
|
16
|
Karl V, Hofmann M, Thimme R. Role of antiviral CD8+ T cell immunity to SARS-CoV-2 infection and vaccination. J Virol 2025; 99:e0135024. [PMID: 40029063 PMCID: PMC11998524 DOI: 10.1128/jvi.01350-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
The COVID-19 pandemic has greatly enhanced our understanding of CD8+ T cell immunity and their role in natural infection and vaccine-induced protection. Rapid and early SARS-CoV-2-specific CD8+ T cell responses have been associated with efficient viral clearance and mild disease. Virus-specific CD8+ T cell responses can compensate for waning, morbidity-related, and iatrogenic reduction of humoral immunity. After infection or vaccination, SARS-CoV-2-specific memory CD8+ T cells are formed, which mount an efficient recall response in the event of breakthrough infection and help to protect from severe disease. Due to their breadth and ability to target mainly highly conserved epitopes, SARS-CoV-2-specific CD8+ T cells are also able to cross-recognize epitopes of viral variants, thus maintaining immunity even after the emergence of viral evolution. In some cases, however, CD8+ T cells may contribute to the pathogenesis of severe COVID-19. In particular, delayed and uncontrolled, e.g., nonspecific and hyperactivated, cytotoxic CD8+ T cell responses have been linked to poor COVID-19 outcomes. In this minireview, we summarize the tremendous knowledge about CD8+ T cell responses to SARS-CoV-2 infection and COVID-19 vaccination that has been gained over the past 5 years, while also highlighting the critical knowledge gaps that remain.
Collapse
Affiliation(s)
- Vivien Karl
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Maike Hofmann
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Thimme
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
17
|
Jennewein MF, Schultz MD, Beaver S, Battisti P, Bakken J, Hanson D, Akther J, Zhou F, Mohamath R, Singh J, Cross N, Kasal DN, Ykema MR, Reed S, Kalange D, Cheatwood IR, Tipper JL, Foote JB, King RG, Silva-Sanchez A, Harrod KS, Botta D, Gerhardt A, Casper C, Randall TD, Lund FE, Voigt EA. Intranasal replicon SARS-CoV-2 vaccine produces protective respiratory and systemic immunity and prevents viral transmission. Mol Ther 2025:S1525-0016(25)00281-3. [PMID: 40211539 DOI: 10.1016/j.ymthe.2025.04.007] [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: 11/04/2024] [Revised: 02/27/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
While mRNA vaccines have been effective in combating SARS-CoV-2, the waning of vaccine-induced antibody responses and lack of vaccine-induced respiratory tract immunity contribute to ongoing infection and transmission. In this work, we compare and contrast intranasal (i.n.) and intramuscular (i.m.) administration of a SARS-CoV-2 replicon vaccine delivered by a nanostructured lipid carrier (NLC). Both i.m. and i.n. vaccines induce potent systemic serum neutralizing antibodies, bone marrow-resident immunoglobulin G-secreting cells, and splenic T cell responses. The i.n. vaccine additionally induces robust respiratory mucosal immune responses, including SARS-CoV-2-reactive lung-resident memory T cell populations. As a booster following previous i.m. vaccination, the i.n. vaccine also elicits the development of mucosal virus-specific T cells. Both the i.m.- and i.n.-administered vaccines durably protect hamsters from infection-associated morbidity upon viral challenge, significantly reducing viral loads and preventing challenged hamsters from transmitting virus to naive cagemates. This replicon-NLC vaccine's potent systemic immunogenicity, and additional mucosal immunogenicity when delivered i.n., may be key for combating SARS-CoV-2 and other respiratory pathogens.
Collapse
Affiliation(s)
- Madeleine F Jennewein
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Michael D Schultz
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Samuel Beaver
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Peter Battisti
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Julie Bakken
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Derek Hanson
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Jobaida Akther
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Fen Zhou
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Raodoh Mohamath
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Jasneet Singh
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Noah Cross
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Darshan N Kasal
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Matthew R Ykema
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Sierra Reed
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Davies Kalange
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Isabella R Cheatwood
- Undergraduate Immunology Program, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer L Tipper
- Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy B Foote
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - R Glenn King
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Aaron Silva-Sanchez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kevin S Harrod
- Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Davide Botta
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Immunology Institute, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alana Gerhardt
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA
| | - Corey Casper
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA; Department of Global Health, University of Washington, Seattle, WA 98195, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Troy D Randall
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Immunology Institute, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Frances E Lund
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Immunology Institute, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emily A Voigt
- Access to Advanced Health Institute (formerly Infectious Disease Research Institute), Seattle, WA 98102, USA.
| |
Collapse
|
18
|
Peters MQ, Young AL, Stolarczuk JE, Glad M, Layton E, Logue JK, Minkah NK, Chu HY, Englund JA, Sather DN, Seshadri C, Kachikis A, Harrington WE. Infant CD4 T-cell response to SARS-CoV-2 mRNA vaccination is restricted in cytokine production and modified by vaccine manufacturer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646864. [PMID: 40271053 PMCID: PMC12016048 DOI: 10.1101/2025.04.02.646864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Safe and effective vaccines are a key preventative measure to protect infants from SARS-CoV-2 infection and disease. Although mRNA vaccines induce robust antibody titers in infants, little is known about the quality of CD4 T-cell responses induced by vaccination. CD4 T-cell responses are important in orchestrating coordinated immune responses during infection and may help to limit disease severity. METHODS To characterize the CD4 T-cell response to SARS-CoV-2 mRNA vaccination in infants, we sampled blood from 13 infants before and after primary SARS-CoV-2 mRNA vaccine series; samples from 12 historical vaccinated adults were used for comparisons. PBMC were stimulated with Spike peptide pools and the ability of CD4 T-cells to secrete Th1, Th2, and Th17 cytokines was quantified. A measure of polyfunctionality was generated using the COMPASS algorithm. RESULTS We observed a significant increase in CD4 T-cells producing IL-2 (0.01% vs. 0.08%, p=0.04) and TNF-α (0.007% vs. 0.07%, p=0.007) following vaccination in infants but a more muted induction of IFN-γ production (0.01% vs 0.04%, p=0.08). This contrasted with adults, in whom vaccination induced robust production of IFN-γ, IL-2, and TNF-α. Th2 and Th17 responses were limited in both infants and adults. In infants, CD4 T-cell responses post-vaccination were greater in those who received mRNA-1273 versus BNT162b. In contrast to CD4 T-cell responses, Spike-specific IgG titers were similar in infants and adults. CONCLUSIONS These data suggest that infants have restricted induction of cytokine producing CD4 T-cells following SARS-CoV-2 mRNA vaccination relative to adults.
Collapse
|
19
|
Yang OO. The immunopathogenesis of SARS-CoV-2 infection: Overview of lessons learned in the first 5 years. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf033. [PMID: 40180332 DOI: 10.1093/jimmun/vkaf033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/11/2025] [Indexed: 04/05/2025]
Abstract
This review provides a broad overview of lessons learned in the five years since COVID-19 was identified. It is a bimodal disease, starting with an initially virus-driven phase, followed by resolution or ensuing inappropriate immune activation causing severe inflammation that is no longer strictly virus dependent. Humoral immunity is beneficial for preventing or attenuating the early stage, without benefit once the later stage begins. Neutralizing antibodies elicited by natural infection or vaccination are short-lived and highly vulnerable to viral sequence variation. By contrast, cellular immunity, particularly the CD8+ T cell arm, has a role in preventing or attenuating severe disease, is far less susceptible to viral variation, and is longer-lived than antibodies. Finally, an ill-defined phenomenon of prolonged symptoms after acute infection, termed "long COVID," is poorly understood but may involve various immunologic defects that are hyperactivating or immunosuppressive. Remaining issues include needing to better understand the immune dysregulation of severe disease to allow more tailored therapeutic interventions, developing antibody strategies that cope with the viral spike sequence variability, prolonging vaccine efficacy, and unraveling the mechanisms of long COVID to design therapeutic approaches.
Collapse
Affiliation(s)
- Otto O Yang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
20
|
Olivieri G, Amodio D, Manno EC, Santilli V, Cotugno N, Palma P. Shielding the immunocompromised: COVID-19 prevention strategies for patients with primary and secondary immunodeficiencies. Vaccine 2025; 51:126853. [PMID: 39946827 DOI: 10.1016/j.vaccine.2025.126853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 03/05/2025]
Abstract
The COVID-19 pandemic has significantly impacted immunocompromised patients, particularly those with inborn errors of immunity (IEI), transplant recipients, hematologic malignancies, and those undergoing treatment with immunosuppressive biologics and medications. These patients face an elevated risk of experiencing severe or even fatal consequences following SARS-CoV-2 infections. Vaccination is the primary defense against COVID-19; however, immune responses following immunization are often suboptimal in these patients, with variable specific humoral response rates. Despite the expedited regulatory approval and the widespread implementation of COVID-19 vaccines, the efficacy and safety for immunocompromised populations require thorough investigation. In future pandemics, including vulnerable populations (VPs) in vaccine and monoclonal antibody (mAb) trials is crucial to develop safe, effective immunization strategies, address gaps in vaccine efficacy and safety data, and create tailored guidelines for at-risk groups. This review provides a comprehensive examination of the efficacy of COVID-19 vaccines and mAbs in patients with primary and secondary immunodeficiency, with a specific focus on individuals with IEI, considering previous regulatory aspects and the necessity of including VPs in vaccine trials to enhance the quality of patient care and promote equitable health outcomes in future pandemics.
Collapse
Affiliation(s)
- Giulio Olivieri
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; PhD Program in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy
| | - Donato Amodio
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Chair of Pediatrics, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Emma Concetta Manno
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Veronica Santilli
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Nicola Cotugno
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Chair of Pediatrics, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Palma
- Chair of Pediatrics, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Centre for the Evaluation of Vaccination and Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
21
|
Dhawan M, Thakur N, Sharma M, Rabaan AA. The comprehensive insights into the B-cells-mediated immune response against COVID-19 infection amid the ongoing evolution of SARS-CoV-2. Biomed Pharmacother 2025; 185:117936. [PMID: 40056829 DOI: 10.1016/j.biopha.2025.117936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 03/10/2025] Open
Abstract
The antibody-mediated immune response is crucial for the development of protective immunity against SARS-CoV-2, the virus responsible for the COVID-19 pandemic. Understanding the interaction between SARS-CoV-2 and the immune system is critical because new variants emerge as a result of the virus's ongoing evolution. Understanding the function of B cells in the SARS-CoV-2 infection process is critical for developing effective and long-lasting vaccines against this virus. Triggered by the innate immune response, B cells transform into memory B cells (MBCs). It is fascinating to observe how MBCs provide enduring immune defence, not only eradicating the infection but also safeguarding against future reinfection. If there is a lack of B cell activation or if the B cells are not functioning properly, it can lead to a serious manifestation of the disease and make immunisation less effective. Individuals with disruptions in the B cells have shown increased production of cytokines and chemokines, resulting in a poor prognosis for the disease. Therefore, we have developed an updated review article to gain insight into the involvement of B cells in SARS-CoV-2 infection. The discussion has covered the generation, functioning, and dynamics of neutralising antibodies (nAbs). Furthermore, we have emphasised immunotherapeutics that rely on nAbs.
Collapse
Affiliation(s)
- Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, Punjab 141004, India; Trafford College, Altrincham, Altrincham, Manchester WA14 5PQ, UK.
| | - Nanamika Thakur
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, India
| | - Manish Sharma
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, India
| | - Ali A Rabaan
- Research Center, Dr. Sulaiman Alhabib Medical Group, Riyadh 13328, Saudi Arabia; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan.
| |
Collapse
|
22
|
Kim AR, Koh JY, Rha MS, Jung JH, Ko JH, Choi HK, Jeon JH, Seok H, Park DW, Peck KR, Choi JY, Park SH, Choi WS, Jeong HW, Shin EC. Patients With Mild COVID-19 Exhibit Low Functional Avidity of SARS-CoV-2 Membrane Protein-Reactive CD4 + T Cells. Immune Netw 2025; 25:e4. [PMID: 40342838 PMCID: PMC12056292 DOI: 10.4110/in.2025.25.e4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/28/2024] [Accepted: 01/06/2025] [Indexed: 05/11/2025] Open
Abstract
Herein, we found that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-unexposed individuals exhibited an increased frequency of CD4+ T cells against SARS-CoV-2 membrane (M) protein, suggesting that SARS-CoV-2 M-reactive cells may be primed by previous infection with common cold coronaviruses (CCCoVs). We confirmed that CCCoV M-reactive CD4+ T cells cross-recognize SARS-CoV-2 M in unexposed individuals. Among coronavirus disease 2019 (COVID-19) convalescents and unexposed individuals, SARS-CoV-2 M-reactive CD4+ T cells exhibited significantly lower functional avidity than CD4+ T cells reactive to other viruses. Importantly, convalescents from mild COVID-19 had SARS-CoV-2 M-reactive CD4+ T cells with significantly lower functional avidity than convalescents from severe COVID-19. The current data suggest that pre-existing CCCoV M-specific memory CD4+ T cells may contribute to controlling SARS-CoV-2 infection by cross-reactivity, leading to mild disease but leaving memory cells with low functional avidity to SARS-CoV-2 M due to incomplete homology. These data provide indirect evidence that pre-existing cross-reactive CD4+ T cells contribute to protection from severe COVID-19.
Collapse
Affiliation(s)
- A-Reum Kim
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - June-Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Min-Seok Rha
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jae Hyung Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jae-Hoon Ko
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hee Kyoung Choi
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan 15355, Korea
| | - Ji Hoon Jeon
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan 15355, Korea
| | - Hyeri Seok
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan 15355, Korea
| | - Dae Won Park
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan 15355, Korea
| | - Kyong Ran Peck
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Jun Yong Choi
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- The Center for Epidemic Preparedness, KAIST, Daejeon 34141, Korea
| | - Won Suk Choi
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan 15355, Korea
| | - Hye Won Jeong
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Eui-Cheol Shin
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
23
|
Dörnte C, Datsi A, Traska V, Kostyra J, Wagner M, Brauns O, Lamsfuß C, Winkels H, Balz V, Enczmann J, Adams O, Mueller L, Baurmann H, Eiz‐Vesper B, Bonifacius A, Sorg RV, Dose C, Schmitz J, Richter A, Fischer J, Schuster M. Distinct HLA Haplotypes Are Associated With an Altered Strength of SARS-CoV-2-Specific T-Cell Responses and Unfavorable Disease Courses. Eur J Immunol 2025; 55:e202451497. [PMID: 40259456 PMCID: PMC12012228 DOI: 10.1002/eji.202451497] [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: 09/03/2024] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 04/23/2025]
Abstract
Infection with SARS-CoV-2 results in mild to severe COVID-19 disease courses. Several studies showed the association of impaired T-cell responses and certain HLA haplotypes with disease severity. However, it remained unclear if T-cell activation was compromised due to a general reduction of presented epitopes or other intrinsic factors within APCs or T cells. Furthermore, a potential reduction of presented epitopes would suggest if an upcoming SARS-CoV-2 variant could escape T-cell immunity. Hence, knowledge about the T-cell epitope landscape of SARS-CoV-2 would allow to better understand mechanisms leading to severe disease and to estimate the potential stability of the T-cell response in light of virus evolution, which might provide insights for future vaccine designs. Hence, in the present study, the T-cell epitope landscape of SARS-CoV-2 was determined via in vitro T-cell stimulation plus in silico prediction. HLAs associated with mild and severe disease courses showed almost the same potential in epitope presentation, suggesting intrinsic factors of APCs or T cells as contributors to the more severe disease courses. As T-cell epitopes did also not originate from regions of SARS-CoV-2 having shown high mutation rates in the past, a relatively stable T-cell response can be expected regarding new SARS-CoV-2 strains in the future. Analysis of the T-cell epitope landscape of SARS-CoV-2 suggests T-cell intrinsic factors as likely modulators of disease severity and that the capacity of MHC-peptide presentation remains stable among circulating SARS-CoV-2 viral strains.
Collapse
Affiliation(s)
- C. Dörnte
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
- Clinic III for Internal MedicineUniversity of Cologne Faculty of Medicine and University Hospital CologneCologneGermany
| | - A. Datsi
- Institute for Transplantation Diagnostics and Cell TherapeuticsMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐University DüsseldorfDüsseldorfGermany
| | - V. Traska
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - J. Kostyra
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - M. Wagner
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - O. Brauns
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - C. Lamsfuß
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - H. Winkels
- Clinic III for Internal MedicineUniversity of Cologne Faculty of Medicine and University Hospital CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - V. Balz
- Institute for Transplantation Diagnostics and Cell TherapeuticsMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐University DüsseldorfDüsseldorfGermany
| | - J. Enczmann
- Institute for Transplantation Diagnostics and Cell TherapeuticsMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐University DüsseldorfDüsseldorfGermany
| | - O. Adams
- Institute for VirologyMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐UniversityDüsseldorfGermany
| | - L. Mueller
- Institute for VirologyMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐UniversityDüsseldorfGermany
| | - H. Baurmann
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - B. Eiz‐Vesper
- Institute of Transfusion Medicine and Transplant EngineeringMedizinische Hochschule HannoverHannoverGermany
| | - A. Bonifacius
- Institute of Transfusion Medicine and Transplant EngineeringMedizinische Hochschule HannoverHannoverGermany
| | - R. V. Sorg
- Institute for Transplantation Diagnostics and Cell TherapeuticsMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐University DüsseldorfDüsseldorfGermany
| | - C. Dose
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - J. Schmitz
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - A. Richter
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - J. Fischer
- Institute for Transplantation Diagnostics and Cell TherapeuticsMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐University DüsseldorfDüsseldorfGermany
| | - M. Schuster
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| |
Collapse
|
24
|
Fu W, Zhang W, You Z, Li G, Wang C, Lei C, Zhao J, Hou J, Hu S. T-Cell-Dependent Bispecific IgGs Protect Aged Mice From Lethal SARS-CoV-2 Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406980. [PMID: 39976129 PMCID: PMC12005765 DOI: 10.1002/advs.202406980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 01/04/2025] [Indexed: 02/21/2025]
Abstract
T-cell ageing may be a key factor in the disproportionate severity of coronavirus disease 2019 (COVID-19) in older populations. For hospitalized COVID-19 patients, treatment involving the use of monoclonal antibodies with the ability to neutralize SARS-CoV-2 usually involves the administration of high doses but has not been very effective at preventing complications or fatality, highlighting the need for additional research into anti-SARS-CoV-2 therapies, particularly for older populations. In this study, it is discovered that older persons with a severe SARS-CoV-2 infection has weaker T-cell responses. Therefore the development and characterization of spike-targeting T-cell-dependent bispecific (TDB) full-length human immunoglobulin Gs with enhanced efficacy in the treatment of COVID-19 is described. Using S-targeting TDBs, polyclonal T cells are guided to target and destroy S-expressing cells, preventing the cell-to-cell transmission of SARS-CoV-2 and thereby eliminating the need for SARS-CoV-2-specific immunity. Using animal models of COVID-19, it is shown that the selective activation of T cells improves the efficiency of treatment in preinfected mice by attenuating disease-induced weight loss and death. The significance of T-cell-based immunity during infection is highlighted by the findings. These results have implications for better clinical effectiveness of therapies for COVID-19 and the development of T-cell-dependent medicines for the elderly population.
Collapse
Affiliation(s)
- Wenyan Fu
- Department of Assisted ReproductionShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Department of Biomedical EngineeringCollege of Basic Medical SciencesSecond Military Medical UniversityShanghai200433China
| | - Wei Zhang
- Department of Respiratory and Critical Care MedicineFirst Affiliated HospitalSecond Military Medical UniversityShanghai200433China
- Center of Critical Care MedicineFirst Affiliated Hospitalthe Second Military Medical UniversityShanghai200433China
| | - Zhongshuai You
- Department of Biomedical EngineeringCollege of Basic Medical SciencesSecond Military Medical UniversityShanghai200433China
| | - Guangyao Li
- Department of BiophysicsCollege of Basic Medical, SciencesSecond Military Medical UniversityShanghai200433China
| | - Chuqi Wang
- Department of Biomedical EngineeringCollege of Basic Medical SciencesSecond Military Medical UniversityShanghai200433China
| | - Changhai Lei
- Department of BiophysicsCollege of Basic Medical, SciencesSecond Military Medical UniversityShanghai200433China
| | - Jian Zhao
- KOCHKOR Biotech, Inc.Shanghai201406China
| | - Jin Hou
- National Key Laboratory of Medical Immunology and Institute of ImmunologySecond Military Medical UniversityShanghai200433China
| | - Shi Hu
- Department of Biomedical EngineeringCollege of Basic Medical SciencesSecond Military Medical UniversityShanghai200433China
| |
Collapse
|
25
|
Spector BL, Koseva B, McLennan R, Banerjee D, Lankachandra K, Bradley T, Selvarangan R, Grundberg E. Methylation patterns of the nasal epigenome of hospitalized SARS-CoV-2 positive patients reveal insights into molecular mechanisms of COVID-19. BMC Med Genomics 2025; 18:62. [PMID: 40170038 PMCID: PMC11963311 DOI: 10.1186/s12920-025-02125-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 03/12/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has varied presentations from asymptomatic to death. Efforts to identify factors responsible for differential COVID-19 severity include but are not limited to genome wide association studies (GWAS) and transcriptomic analysis. More recently, variability in host epigenomic profiles have garnered attention, providing links to disease severity. However, whole epigenome analysis of the respiratory tract, the target tissue of SARS-CoV-2, remains ill-defined. RESULTS We interrogated the nasal methylome to identify pathophysiologic drivers in COVID-19 severity through whole genome bisulfite sequencing (WGBS) of nasal samples from COVID-19 positive individuals with severe and mild presentation of disease. We noted differential DNA methylation in intergenic regions and low methylated regions (LMRs), demonstrating the importance of distal regulatory elements in gene regulation in COVID-19 illness. Additionally, we demonstrated differential methylation of pathways implicated in immune cell recruitment and function, and the inflammatory response. We found significant hypermethylation of the FUT4 promoter implicating impaired neutrophil adhesion in severe disease. We also identified hypermethylation of ELF5 binding sites suggesting downregulation of ELF5 targets in the nasal cavity as a factor in COVID-19 phenotypic variability. CONCLUSIONS This study demonstrated DNA methylation as a marker of the immune response to SARS-CoV-2 infection, with enhancer-like elements playing significant roles. It is difficult to discern whether this differential methylation is a predisposing factor to severe COVID-19, or if methylation differences occur in response to disease severity. These differences in the nasal methylome may contribute to disease severity, or conversely, the nasal immune system may respond to severe infection through differential immune cell recruitment and immune function, and through differential regulation of the inflammatory response.
Collapse
Affiliation(s)
- Benjamin L Spector
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI, 53792, USA.
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA.
| | - Boryana Koseva
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Rebecca McLennan
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Dithi Banerjee
- Department of Pathology and Laboratory Medicine, Children'S Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Kamani Lankachandra
- Department of Pathology, University Health, University of Missouri- Kansas City School of Medicine, 2411 Holmes St, Kansas City, MO, 64108, USA
| | - Todd Bradley
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Rangaraj Selvarangan
- Department of Pathology and Laboratory Medicine, Children'S Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Elin Grundberg
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA.
| |
Collapse
|
26
|
Augello M, Bono V, Rovito R, Tavelli A, Santoro A, Tincati C, Vergori A, Azzini AM, Righi E, Spiteri G, Porru S, Meschi S, Notari S, Maggi F, Antinori A, Tacconelli E, d'Arminio Monforte A, Marchetti G. Long-term immune responses to SARS-CoV-2 Omicron BA.4/5 mRNA booster in people living with HIV. COMMUNICATIONS MEDICINE 2025; 5:92. [PMID: 40148493 PMCID: PMC11950219 DOI: 10.1038/s43856-025-00799-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Variant-adapted vaccines are recommended in vulnerable populations to address the waning immunity and the emergence of immune-escaping SARS-CoV-2 variants, yet data about immune responses to such vaccines in people living with HIV (PLWH) are limited. We therefore aimed to assess long-term immune responses to an original-BA.4/5 mRNA booster in this population. METHODS In this prospective longitudinal study, PLWH receiving either an original-BA.4/5 bivalent booster or an original monovalent booster and HIV-negative healthcare workers (HCWs) receiving a bivalent booster were enrolled and sampled before (T0), 1 month (T1), and 4-9 months (T2) after the vaccine administration. SARS-CoV-2-specific T and B cells, RBD-binding antibodies, and RBD-blocking antibodies against both wild type (WT) and omicron BA.4/5 virus were determined. RESULTS The bivalent booster is able to transiently increase both humoral and polyfunctional T cell responses in PLWH, with humoral responses comparable to those observed in HCWs. While T cell responses are cross-reactive against viral variants and stable over time, humoral immunity is imprinted to the ancestral virus and wanes quickly. Furthermore, whilst previous SARS-CoV-2 infection does not affect the trajectory of vaccine-elicited immune responses, markers of HIV-related T cell dysfunction are associated with lower antibody peak responses and higher antibody waning. Lastly, the bivalent booster was superior to the monovalent one in inducing BA.4/5-reactive RBD-blocking antibodies. CONCLUSIONS The original-BA.4/5 bivalent booster is highly immunogenic in PLWH and superior to the monovalent one in inducing humoral responses against the BA.4/5 virus, although HIV-related T cell dysfunction markers are associated with blunted and less durable antibody immunity.
Collapse
Affiliation(s)
- Matteo Augello
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Valeria Bono
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Roberta Rovito
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | | | - Andrea Santoro
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Camilla Tincati
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Alessandra Vergori
- Viral Immunodeficiencies Unit, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Anna Maria Azzini
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Elda Righi
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Gianluca Spiteri
- Occupational Medicine Unit, Verona Hospital, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Stefano Porru
- Occupational Medicine Unit, Verona Hospital, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Silvia Meschi
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Stefania Notari
- Laboratory of Cellular Immunology and Pharmacology, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Andrea Antinori
- Viral Immunodeficiencies Unit, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Evelina Tacconelli
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Giulia Marchetti
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
27
|
Hsu CY, Chiu CH, Lin TY, Chang FY. Severe SARS-CoV-2 alpha variant convalescent patients exhibit worse T cell immune response than those with mild severity disease. J Formos Med Assoc 2025:S0929-6646(25)00110-X. [PMID: 40155285 DOI: 10.1016/j.jfma.2025.03.009] [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: 10/04/2024] [Revised: 01/14/2025] [Accepted: 03/07/2025] [Indexed: 04/01/2025] Open
Abstract
OBJECTIVES This study aimed to assess T cell phenotype and the role of cellular immunity against acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants in alpha variant coronavirus disease 2019 (COVID-19) convalescent patients. METHODS Thirty-two confirmed SARS-CoV-2 infected patients and ten healthy controls were enrolled. T cell subsets in peripheral blood were classified and quantified using flow cytometry. Additionally, T cell immune responses against SARS-CoV-2 spike peptide pools were assessed. Flow cytometry data were analyzed using Cytobank software. Other analyses involved Student's t-test or chi-square test. RESULTS CD127 expression on T helper cells and cytotoxic T cells was lower in the severe disease group than that in the mild disease group. Severe COVID-19 convalescents with SARS-CoV-2 alpha variant exhibited poorer T cell immune responses than those with mild disease upon spike peptide pools stimulation with SARS-CoV-2 wild type, alpha, or omicron variants. CONCLUSIONS COVID-19 convalescents showed T helper and cytotoxic T cells with lower CD127 expression in the severe disease group than those in the mild disease group. Severe COVID-19 convalescents infected with the alpha variant exhibited poorer T cell immune responses against the SARS-CoV-2 wild type, alpha, or omicron variants. Our study provides insights into the differences in T cell phenotypes and immune responses during the contraction phase following SARS-CoV-2 infection across varying disease severities. These findings offer valuable perspectives for advancing future research on SARS-CoV-2 T cell-related immune responses.
Collapse
Affiliation(s)
- Chih-Yao Hsu
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Hsiang Chiu
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Te-Yu Lin
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Feng-Yee Chang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
28
|
Mackay M, Wagner CA, Pinckney A, Cohen JA, Wallace ZS, Khosroshahi A, Sparks JA, Lord S, Saxena A, Caricchio R, Kim AH, Kamen DL, Koumpouras F, Askanase AD, Smith K, Guthridge JM, Pardo G, Mao-Draayer Y, Macwana S, McCarthy S, Sherman MA, Hamrah SD, Veri M, Walker S, York K, Tedeschi S, Wang J, Dziubla G, Castro M, Carroll R, Narpala S, Lin BC, Serebryannyy L, McDermott A, Barry WT, Goldmuntz E, McNamara J, Payne AS, Bar-Or A, Khanna D, James JA. Prospective SARS-CoV-2 Booster Vaccination in Immunosuppressant-Treated Systemic Autoimmune Disease Patients in a Randomized Controlled Trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.25.25324558. [PMID: 40196267 PMCID: PMC11974989 DOI: 10.1101/2025.03.25.25324558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background Autoimmune disease patients on immunosuppressants exhibit reduced humoral responses to primary COVID-19 vaccination. Booster vaccine responses and the effects of holding immunosuppression around vaccination are less studied. We evaluated the efficacy and safety of additional vaccination in mycophenolate mofetil/mycophenolic acid (MMF/MPA)-, methotrexate (MTX)-, and B cell-depleting therapy (BCDT)-treated autoimmune disease patients, including the impact of withholding MMF/MPA and MTX. Methods In this open-label, multicenter, randomized trial, 22 MMF/MPA-, 26 MTX-, and 93 BCDT-treated autoimmune disease patients with negative or suboptimal antibody responses to initial COVID-19 vaccines (BNT162b2, mRNA-1273, or AD26.COV2.S) received a homologous booster. MMF/MPA and MTX participants were randomized (1:1) to continue or withhold treatment around vaccination. The primary outcome was the change in anti-Wuhan-Hu-1 receptor-binding domain (RBD) concentrations at 4 weeks post-additional vaccination. Secondary outcomes included adverse events, COVID-19 infections, and autoimmune disease activity through 48 weeks. Results Additional vaccination increased anti-RBD concentrations in MMF/MPA and MTX patients, irrespective of whether immunosuppression was continued or withheld. BCDT-treated patients also demonstrated increased anti-RBD concentrations, albeit lower than MMF/MPA- and MTX-treated cohorts. COVID-19 infections occurred in 30-46% of participants, were predominantly mild, and included only two non-fatal hospitalizations. Additional vaccination was well-tolerated, with low frequencies of severe disease flares and adverse events. Conclusion Additional COVID-19 vaccination is effective and safe in immunosuppressant-treated autoimmune disease patients, regardless of whether MMF/MPA or MTX is withheld. Trial Registration. ClinicalTrials.gov (NCT#05000216).
Collapse
|
29
|
Samaan P, Korosec CS, Budylowski P, Chau SLL, Pasculescu A, Qi F, Delgado-Brand M, Tursun TR, Mailhot G, Dayam RM, Arnold CR, Langlois MA, Mendoza J, Morningstar T, Law R, Mihelic E, Sheikh-Mohamed S, Cao EY, Paul N, Patel A, de Launay KQ, Boyd JM, Takaoka A, Colwill K, Matveev V, Yue FY, McGeer A, Straus S, Gingras AC, Heffernen JM, Ostrowski M. mRNA vaccine-induced SARS-CoV-2 spike-specific IFN-γ and IL-2 T-cell responses are predictive of serological neutralization and are transiently enhanced by pre-existing cross-reactive immunity. J Virol 2025; 99:e0168524. [PMID: 39887249 PMCID: PMC11915849 DOI: 10.1128/jvi.01685-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 02/01/2025] Open
Abstract
The contributions of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells to vaccine efficacy and durability are unclear. We investigated relationships between mRNA vaccine-induced spike-specific interferon- gamma (IFN-γ) and interleukin-2 (IL-2) T-cell responses and neutralizing antibody development in long-term care home staff doubly vaccinated with BNT162b2 or mRNA-1273. The impacts of pre-existing cross-reactive T-cell immunity on cellular and humoral responses to vaccination were additionally assessed. Mathematical modeling of the kinetics of spike-specific IFN-γ and IL-2 T-cell responses over 6 months post-second dose was bifurcated into recipients who exhibited gradual increases with doubling times of 155 and 167 days or decreases with half-lives of 165 and 132 days, respectively. Differences in kinetics did not correlate with clinical phenotypes. Serological anti-spike IgG, anti-receptor binding domain (RBD) IgG, anti-spike IgA, and anti-RBD IgA antibody levels otherwise decayed in all participants with half-lives of 63, 57, 79, and 46 days, respectively, alongside waning neutralizing capacity (t1/2 = 408 days). Spike-specific T-cell responses induced at 2-6 weeks positively correlated with live viral neutralization at 6 months post-second dose, especially in hybrid immune individuals. Participants with pre-existing cross-reactive T-cell immunity to SARS-CoV-2 exhibited greater spike-specific T-cell responses, reduced anti-RBD IgA antibody levels, and a trending increase in neutralization at 2-6 weeks post-second dose. Non-spike-specific T-cells predominantly targeted SARS-CoV-2 non-structural protein at 6 months post-second dose in cross-reactive participants. mRNA vaccination was lastly shown to induce off-target T-cell responses against unrelated antigens. In summary, vaccine-induced spike-specific T-cell immunity appeared to influence serological neutralizing capacity, with only a modest effect induced by pre-existing cross-reactivity. IMPORTANCE Our findings provide valuable insights into the potential contributions of mRNA vaccine-induced spike-specific T-cell responses to the durability of neutralizing antibody levels in both uninfected and hybrid immune recipients. Our study additionally sheds light on the precise impacts of pre-existing cross-reactive T-cell immunity to severe acute respiratory syndrome coronavirus 2 on the magnitude and kinetics of cellular and humoral responses to vaccination. Accordingly, our data will help optimize the development of next-generation T cell-based coronavirus vaccines and vaccine regimens to maximize efficacy and durability.
Collapse
Affiliation(s)
- Philip Samaan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Chapin S. Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, Toronto, Ontario, Canada
- Center for Disease Modelling, Mathematics and Statistics, York University, Toronto, Ontario, Canada
| | - Patrick Budylowski
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Serena L. L. Chau
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Pasculescu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Freda Qi
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | | | - Tulunay R. Tursun
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Geneviève Mailhot
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Roya Monica Dayam
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Corey R. Arnold
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Justin Mendoza
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Ryan Law
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Erik Mihelic
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Eric Yixiao Cao
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Nimitha Paul
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Anjali Patel
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | | | - Jamie M. Boyd
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Alyson Takaoka
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Vitaliy Matveev
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Feng Yun Yue
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Allison McGeer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Sharon Straus
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jane M. Heffernen
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, Toronto, Ontario, Canada
- Center for Disease Modelling, Mathematics and Statistics, York University, Toronto, Ontario, Canada
| | - Mario Ostrowski
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
- Keenan Research Center for Biomedical Science, St Michael's Hospital Keenan, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Majumdar S, Weaver JD, Pontejo SM, Minai M, Lu X, Gao JL, Holmes G, Johnson R, Zhang H, Kelsall BL, Farber JM, Alves DA, Murphy PM. Cxcl10 is protective during mouse-adapted SARS-CoV-2 infection. J Leukoc Biol 2025; 117:qiae252. [PMID: 39607906 PMCID: PMC11953068 DOI: 10.1093/jleuko/qiae252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 11/30/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent of the coronavirus disease 2019 (COVID-19) pandemic, remains endemic worldwide. Circulating levels of the chemokine CXCL10 are strongly positively associated with poor outcome; however, its precise role in SARS-CoV-2 pathogenesis and its suitability as a therapeutic target have remained undefined. Here, we challenged mice genetically deficient in Cxcl10 with a mouse-adapted strain of SARS-CoV-2. Infected male, but not female, Cxcl10-/- mice displayed increased mortality compared to wild type controls. Histopathological damage, inflammatory gene induction, and virus load in the lungs of male mice were not broadly influenced by Cxcl10 deficiency. However, accumulation of B and T lymphocytes in the lung parenchyma of infected mice was reduced in the absence of Cxcl10. Thus, during acute SARS-CoV-2 infection, Cxcl10 regulates lymphocyte infiltration in lung and confers protection against mortality. Our preclinical model results do not support targeting CXCL10 therapeutically in severe COVID-19.
Collapse
Affiliation(s)
- Shamik Majumdar
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Joseph D Weaver
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Sergio M Pontejo
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, United States
| | - Xinping Lu
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Ji-Liang Gao
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Gibran Holmes
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Reed Johnson
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Hongwei Zhang
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Brian L Kelsall
- Mucosal Immunobiology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Joshua M Farber
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Derron A Alves
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, United States
| | - Philip M Murphy
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| |
Collapse
|
31
|
Guercetti J, Alorda M, Sappia L, Galve R, Duran-Corbera M, Pulido D, Berardi G, Royo M, Lacoma A, Muñoz J, Padilla E, Castañeda S, Sendra E, Horcajada JP, Gutierrez-Galvez A, Marco S, Salvador JP, Marco MP. Immuno-μSARS2 Chip: A Peptide-Based Microarray to Assess COVID-19 Prognosis Based on Immunological Fingerprints. ACS Pharmacol Transl Sci 2025; 8:871-884. [PMID: 40109734 PMCID: PMC11915183 DOI: 10.1021/acsptsci.4c00727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
A multiplexed microarray chip (Immuno-μSARS2) aiming at providing information on the prognosis of the COVID-19 has been developed. The diagnostic technology records information related to the profile of the immunological response of patients infected by the SARS-CoV-2 virus. The diagnostic technology delivers information on the avidity of the sera against 28 different peptide epitopes and 7 proteins printed on a 25 mm2 area of a glass slide. The peptide epitopes (12-15 mer) derived from structural proteins (Spike and Nucleocapsid) have been rationally designed, synthesized, and used to develop Immuno-μSARS2 as a multiplexed and high-throughput fluorescent microarray platform. The analysis of 755 human serum samples (321 from PCR+ patients; 288 from PCR- patients; 115 from prepandemic individuals and classified as hospitalized, admitted to intensive-care unit (ICU), and exitus) from three independent cohorts has shown that the chips perform with a 98% specificity and 91% sensitivity identifying RT-PCR+ patients. Computational analysis utilized to correlate the immunological signatures of the samples analyzed indicate significant prediction rates against exitus conditions with 82% accuracy, ICU admissions with 80% accuracy, and 73% accuracy over hospitalization requirement compared to asymptomatic patients' fingerprints. The miniaturized microarray chip allows simultaneous determination of 96 samples (24 samples/slide) in 90 min and requires only 10 μL of sera. The diagnostic approach presented for the first time here could have a great value in assisting clinicians in decision-making based on the information provided by the Immuno-μSARS2 regarding progression of the disease and could be easily implemented in diagnostics of other infectious diseases.
Collapse
Affiliation(s)
- Julian Guercetti
- Nanobiotechnology for Diagnostics Group, Instituto de Química Avanzada de Cataluña, IQAC-CSIC, C/Jordi Girona 18-26, 08034 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marc Alorda
- Nanobiotechnology for Diagnostics Group, Instituto de Química Avanzada de Cataluña, IQAC-CSIC, C/Jordi Girona 18-26, 08034 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Marti i Franqués 1-11, 08028 Barcelona, Spain
| | - Luciano Sappia
- Nanobiotechnology for Diagnostics Group, Instituto de Química Avanzada de Cataluña, IQAC-CSIC, C/Jordi Girona 18-26, 08034 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Roger Galve
- Nanobiotechnology for Diagnostics Group, Instituto de Química Avanzada de Cataluña, IQAC-CSIC, C/Jordi Girona 18-26, 08034 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Macarena Duran-Corbera
- Multivalent Systems for Nanomedicine (MS4N), Instituto de Química Avanzada de Cataluña, IQAC-CSIC, C/Jordi Girona 18-26, 08034 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Pulido
- Multivalent Systems for Nanomedicine (MS4N), Instituto de Química Avanzada de Cataluña, IQAC-CSIC, C/Jordi Girona 18-26, 08034 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ginevra Berardi
- Multivalent Systems for Nanomedicine (MS4N), Instituto de Química Avanzada de Cataluña, IQAC-CSIC, C/Jordi Girona 18-26, 08034 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miriam Royo
- Multivalent Systems for Nanomedicine (MS4N), Instituto de Química Avanzada de Cataluña, IQAC-CSIC, C/Jordi Girona 18-26, 08034 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alicia Lacoma
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut Germans Trias i Pujol, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Muñoz
- Servicio de Microbiología del Laboratorio de Referencia de Catalunya, 08820 Barcelona, Spain
| | - Eduardo Padilla
- Servicio de Microbiología del Laboratorio de Referencia de Catalunya, 08820 Barcelona, Spain
| | - Silvia Castañeda
- Servicio de Enfermedades Infecciosas del Hospital del Mar de Barcelona, COVID-MAR group, 08003 Barcelona, Spain
| | - Elena Sendra
- Servicio de Enfermedades Infecciosas del Hospital del Mar de Barcelona, COVID-MAR group, 08003 Barcelona, Spain
| | - Juan P Horcajada
- Servicio de Enfermedades Infecciosas del Hospital del Mar de Barcelona, COVID-MAR group, 08003 Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Agustín Gutierrez-Galvez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Marti i Franqués 1-11, 08028 Barcelona, Spain
| | - Santiago Marco
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Marti i Franqués 1-11, 08028 Barcelona, Spain
| | - J-Pablo Salvador
- Nanobiotechnology for Diagnostics Group, Instituto de Química Avanzada de Cataluña, IQAC-CSIC, C/Jordi Girona 18-26, 08034 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - M-Pilar Marco
- Nanobiotechnology for Diagnostics Group, Instituto de Química Avanzada de Cataluña, IQAC-CSIC, C/Jordi Girona 18-26, 08034 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
32
|
Hurst JH, Mohan AA, Dalapati T, George IA, Aquino JN, Lugo DJ, Pfeiffer TS, Rodriguez J, Rotta AT, Turner NA, Burke TW, McClain MT, Henao R, DeMarco CT, Louzao R, Denny TN, Walsh KM, Xu Z, Mejias A, Ramilo O, Woods CW, Kelly MS. Age-associated differences in mucosal and systemic host responses to SARS-CoV-2 infection. Nat Commun 2025; 16:2383. [PMID: 40064870 PMCID: PMC11894178 DOI: 10.1038/s41467-025-57655-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Age is among the strongest risk factors for severe outcomes from SARS-CoV-2 infection. Here we describe upper respiratory tract (URT) and peripheral blood transcriptomes of 202 participants (age range of 1 week to 83 years), including 137 non-hospitalized individuals with mild SARS-CoV-2 infection and 65 healthy individuals. Among healthy children and adolescents, younger age is associated with higher URT expression of innate and adaptive immune pathways. SARS-CoV-2 infection induces broad upregulation of URT innate and adaptive immune responses among children and adolescents. Peripheral blood responses among SARS-CoV-2-infected children and adolescents are dominated by interferon pathways, while upregulation of myeloid activation, inflammatory, and coagulation pathways is observed only in adults. Among SARS-CoV-2-infected individuals, fever is associated with blunted URT immune responses and more pronounced systemic immune activation. These findings demonstrate that immune responses to SARS-CoV-2 differ across the lifespan, from distinct signatures in childhood and adolescence to age-associated alterations in adults.
Collapse
Affiliation(s)
- Jillian H Hurst
- Department of Pediatrics, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Children's Health and Discovery Institute, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Aditya A Mohan
- Department of Biomedical Engineering, Duke University School of Medicine, Durham, NC, USA
| | - Trisha Dalapati
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Ian A George
- Duke University School of Medicine, Durham, NC, USA
| | - Jhoanna N Aquino
- Department of Pediatrics, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Debra J Lugo
- Department of Pediatrics, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Trevor S Pfeiffer
- Department of Pediatrics, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Javier Rodriguez
- Children's Clinical Research Unit, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Alexandre T Rotta
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Nicholas A Turner
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Thomas W Burke
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Center for Infectious Disease Diagnostics and Innovation, Duke University School of Medicine, Durham, NC, USA
| | - Micah T McClain
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Center for Infectious Disease Diagnostics and Innovation, Duke University School of Medicine, Durham, NC, USA
- Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Ricardo Henao
- Department of Biostatistics and Informatics, Duke University, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - C Todd DeMarco
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Raul Louzao
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Thomas N Denny
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kyle M Walsh
- Children's Health and Discovery Institute, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Zhaohui Xu
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Asuncion Mejias
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Octavio Ramilo
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Christopher W Woods
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Center for Infectious Disease Diagnostics and Innovation, Duke University School of Medicine, Durham, NC, USA
- Durham Veterans Affairs Medical Center, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Matthew S Kelly
- Department of Pediatrics, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
33
|
Yilmaz IC, Ipekoglu EM, Golcuklu BS, Bildik T, Aksoy AGB, Evcili I, Turay N, Surucu N, Bulbul A, Guvencli N, Yildirim M, Canavar Yildirim T, Atalay YA, Abras I, Ceylan Y, Ozsurekci Y, Tigen ET, Korten V, Gursel M, Gursel I. A phase I/II study of CpG/alum-adjuvanted mammalian-derived quadruple antigen carrying virus-like particle COVID-19 vaccine. Vaccine 2025; 49:126787. [PMID: 39892108 DOI: 10.1016/j.vaccine.2025.126787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Waning Spike-elicited immunity and emerging COVID-19 variants underscore the need for vaccines leveraging multiple SARS-CoV-2 antigens, rapidly adaptable to evolving strains. Herein, we evaluated the safety and immunogenicity of a CpG ODN-adjuvanted, alum-adsorbed, virus-like particle (VLP) vaccine displaying the hexaproline stabilized Spike (S) protein and the Nucleocapsid, Membrane, and Envelope proteins of SARS-CoV-2. METHODS In phase 1 randomized, double-blind, placebo-controlled, dose-escalation trial, participants (N = 38, aged 18-59) received two subcutaneous injections of either 10 μg or 40 μg of VLP or placebo, 21 days apart. The primary and secondary objectives of the study was to evaluate the safety, reactogenicity and immunogenicity, respectively. In the double blind, multi-center phase-2 study, participants (N = 349, aged 18-55) were randomized into three cohorts receiving two doses of 40 μg VLPs displaying Wuhan-Spike, Alpha-Spike, or a combination. The primary and secondary objectives were humoral, and cell mediated immunogenicity (CMI) and safety, respectively. Antibody responses were analyzed using ELISA while ELIspot and CBA assays were used to assess the CMI. RESULTS The VLP vaccine demonstrated a good safety profile, with 255 non-serious adverse events in phase 1 and 308 in phase 2. Five serious AEs were reported in phase 2, all of which were resolved completely. The VLP vaccine, in phase 2, was well-tolerated, elicited moderate but sustained anti-S and anti-N antibody titers for 180 days and induced T-helper-1 biased cellular responses in participants. CONCLUSIONS The VLP platform is rapidly adaptable to accommodate stabilized Spike proteins from emerging variants and inclusion of other structural SARS-CoV-2 proteins could broaden the breadth of T cell-mediated immunity. CLINICALTRIALS gov; NCT04818281 and NCT04962893.
Collapse
MESH Headings
- Humans
- Adult
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/adverse effects
- Female
- Male
- Middle Aged
- COVID-19/prevention & control
- COVID-19/immunology
- Double-Blind Method
- Oligodeoxyribonucleotides/administration & dosage
- SARS-CoV-2/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Young Adult
- Adolescent
- Spike Glycoprotein, Coronavirus/immunology
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/adverse effects
- Adjuvants, Immunologic/administration & dosage
- Alum Compounds/administration & dosage
- Immunogenicity, Vaccine
- Antibodies, Neutralizing/blood
- Adjuvants, Vaccine/administration & dosage
Collapse
Affiliation(s)
- Ismail Cem Yilmaz
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye; Middle East Technical University, Department of Biological Sciences, Ankara, Turkiye
| | - Emre Mert Ipekoglu
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkiye
| | | | - Tugce Bildik
- Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | | | - Irem Evcili
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye; Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | - Nilsu Turay
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye
| | - Naz Surucu
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkiye
| | - Artun Bulbul
- Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | - Nese Guvencli
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkiye
| | - Muzaffer Yildirim
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye
| | - Tugce Canavar Yildirim
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye; Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | - Yagmur Aydin Atalay
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkiye
| | - Irem Abras
- Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | - Yasemin Ceylan
- Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | - Yasemin Ozsurekci
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye; Hacettepe University, Faculty of Medicine, Department of Pediatric Infectious Diseases, Hacettepe University, Faculty of Medicine, Ankara, Turkiye
| | - Elif Tukenmez Tigen
- Marmara University, Infectious Disease and Clinical Microbiology Department, Istanbul, Turkiye
| | - Volkan Korten
- Marmara University, Infectious Disease and Clinical Microbiology Department, Istanbul, Turkiye
| | - Mayda Gursel
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye.
| | - Ihsan Gursel
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye.
| |
Collapse
|
34
|
Fujitani M, Lu X, Shinnakasu R, Inoue T, Kidani Y, Seki NM, Ishida S, Mitsuki S, Ishihara T, Aoki M, Suzuki A, Takahashi K, Takayama M, Ota T, Iwata S, Shibata RY, Sonoyama T, Ariyasu M, Kitano A, Terooatea T, Kelly Villa J, Yamashita K, Yamasaki S, Kurosaki T, Omoto S. Longitudinal analysis of immune responses to SARS-CoV-2 recombinant vaccine S-268019-b in phase 1/2 prime-boost study. Front Immunol 2025; 16:1550279. [PMID: 40109335 PMCID: PMC11919840 DOI: 10.3389/fimmu.2025.1550279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
Background The durability of vaccine-induced immune memory to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is crucial for preventing infection, especially severe disease. Methods This follow-up report from a phase 1/2 study of S-268019-b (a recombinant spike protein vaccine) after homologous booster vaccination confirms its long-term safety, tolerability, and immunogenicity. Results Booster vaccination with S-268019-b resulted in an enhancement of serum neutralizing antibody (NAb) titers and a broad range of viral neutralization. Single-cell immune profiling revealed persistent and mature antigen-specific memory B cells and T follicular helper cells, with increased B-cell receptor diversity. The expansion of B- and T-cell repertoires and presence of cross-reactive NAbs targeting conserved epitopes within the receptor-binding domain following a booster accounted for the broad-spectrum neutralizing activity. Conclusion These findings highlight the potential of S-268019-b to provide broad and robust protection against a range of SARS-CoV-2 variants, addressing a critical challenge in the ongoing fight against coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Masaya Fujitani
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Xiuyuan Lu
- Laboratory of Molecular Immunology, World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Ryo Shinnakasu
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yujiro Kidani
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Naomi M. Seki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Satoru Ishida
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Shungo Mitsuki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | | | - Miwa Aoki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Akio Suzuki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Koji Takahashi
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Masahiro Takayama
- Pharmaceutical Technology Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Takeshi Ota
- Pharmaceutical Technology Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Satoshi Iwata
- Department of Microbiology, Tokyo Medical University, Tokyo, Japan
| | - Risa Yokokawa Shibata
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Takuhiro Sonoyama
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Mari Ariyasu
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | | | | | | | | | - Sho Yamasaki
- Laboratory of Molecular Immunology, World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - Shinya Omoto
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| |
Collapse
|
35
|
Zhu A, Chen Z, Yan Q, Jiang M, Liu X, Li Z, Li N, Tang C, Jian W, He J, Chen L, Cheng J, Chen C, Tang T, Xu Z, Hu Q, Li F, Wang Y, Sun J, Zhuang Z, Wen L, Zhuo J, Liu D, Zhang Y, Huang X, Li S, Zeng Q, Chen F, Zhou L, Liu D, Zhong C, Chen Y, Li S, Liang K, Zhong N, Zhang X, Chen J, Chen X, Xu Y, Zhong N, Zhao J, Zhao J. Robust mucosal SARS-CoV-2-specific T cells effectively combat COVID-19 and establish polyfunctional resident memory in patient lungs. Nat Immunol 2025; 26:459-472. [PMID: 39875584 PMCID: PMC11876067 DOI: 10.1038/s41590-024-02072-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025]
Abstract
Mucosal antigen-specific T cells are pivotal for pathogen clearance and immune modulation in respiratory infections. Dysregulated T cell responses exacerbate coronavirus disease 2019 severity, marked by cytokine storms and respiratory failure. Despite extensive description in peripheral blood, the characteristics of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells in the lungs remain elusive. Here we conducted integrated single-cell profiling of SARS-CoV-2-specific T cells in 122 bronchoalveolar lavage fluid (BALF) and 280 blood samples from 159 patients, including 27 paired BALF and blood samples from 24 patients. SARS-CoV-2-specific T cells were robustly elicited in BALF irrespective of prior vaccination, correlating with diminished viral loads, lessened systemic inflammation and improved respiratory function. SARS-CoV-2-specific T cells in BALF exhibited profound activation, along with proliferative and multi-cytokine-producing capabilities and a glycolysis-driven metabolic signature, which were distinct from those observed in peripheral blood mononuclear cells. After viral clearance, these specific T cells maintained a polyfunctional tissue-resident memory phenotype, highlighting their critical roles in infection control and long-term protection.
Collapse
Grants
- the National Key R&D Program of China (2023YFC2306400 to JC.Z., 2023YFC3041700 to JC.Z.), R&D Program of Guangzhou Laboratory (SRPG22-006 to JC.Z.), National Natural Science Foundation of China (82495200 to JC.Z., 82495203 to JC.Z., 82025001 to JC.Z.)
- R&D Program of Guangzhou Laboratory (EKPG21-29 to A.Z.),National Natural Science Foundation of China (82201933 to A.Z.), China Postdoctoral Science Foundation (2022M710892 to A.Z.), State Key Laboratory of Respiratory Diseases Funded Program (SKLRD-Z-202330 to A.Z.).
- National Natural Science Foundation of China (82201932 to Q.Y.),China Postdoctoral Science Foundation (2022M710891 to Q.Y.), State Key Laboratory of Respiratory Diseases Funded Program (SKLRD-Z-202324 to Q.Y).
- State Key Laboratory of Respiratory Diseases Funded Program (SKLRD-Z-202304 to Q.H.).
- National Key R&D Program of China (2021YFC0864500 to Y.X.), R&D Program of Guangzhou Laboratory (SRPG23-001 to Y.X.)
- Guangdong Basic and Applied Basic Research Foundation (2021B1515130005 to JX.Z.)
Collapse
Affiliation(s)
- Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qihong Yan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mei Jiang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuesong Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhengtu Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Na Li
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Chunli Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenhua Jian
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Lan Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Jinling Cheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Canjie Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tian Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiwei Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingtao Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Fang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhen Zhuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liyan Wen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianfen Zhuo
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Donglan Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanjun Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaofang Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Suxiang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiuhui Zeng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fangli Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liang Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dongdong Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Changhao Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yu Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shiyue Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kangli Liang
- Guangdong Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, China
| | - Na Zhong
- Shenzhen Peacock Biotechnology Co. Ltd, Shenzhen, China
| | - Xinmei Zhang
- Shenzhen Peacock Biotechnology Co. Ltd, Shenzhen, China
| | - Jiekai Chen
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaobo Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Yonghao Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
36
|
van den Dijssel J, Konijn VAL, Duurland MC, de Jongh R, Koets L, Veldhuisen B, Raaphorst H, Turksma AW, Freen‐van Heeren JJ, Steenhuis M, Rispens T, van der Schoot CE, van Ham SM, van Lier RAW, van Gisbergen KPJM, ten Brinke A, van de Sandt CE. Age and Latent Cytomegalovirus Infection Do Not Affect the Magnitude of De Novo SARS-CoV-2-Specific CD8 + T Cell Responses. Eur J Immunol 2025; 55:e202451565. [PMID: 40071711 PMCID: PMC11898545 DOI: 10.1002/eji.202451565] [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: 10/10/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 03/15/2025]
Abstract
Immunosenescence, age-related immune dysregulation, reduces immunity upon vaccinations and infections. Cytomegalovirus (CMV) infection results in declining naïve (Tnaïve) and increasing terminally differentiated (Temra) T cell populations, further aggravating immune aging. Both immunosenescence and CMV have been speculated to hamper the formation of protective T-cell immunity against novel or emerging pathogens. The SARS-CoV-2 pandemic presented a unique opportunity to examine the impact of age and/or CMV on the generation of de novo SARS-CoV-2-specific CD8+ T cell responses in 40 younger (22-40 years) and 37 older (50-66 years) convalescent individuals. Heterotetramer combinatorial coding combined with phenotypic markers were used to study 35 SARS-CoV-2 epitope-specific CD8+ T cell populations directly ex vivo. Neither age nor CMV affected SARS-CoV-2-specific CD8+ T cell frequencies, despite reduced total CD8+ Tnaïve cells in older CMV- and CMV+ individuals. Robust SARS-CoV-2-specific central memory CD8+ T (Tcm) responses were detected in younger and older adults regardless of CMV status. Our data demonstrate that immune aging and CMV status did not impact the SARS-CoV-2-specific CD8+ T cell response. However, SARS-CoV-2-specific CD8+ T cells of older CMV- individuals displayed the lowest stem cell memory (Tscm), highest Temra and PD1+ populations, suggesting that age, not CMV, may impact long-term SARS-CoV-2 immunity.
Collapse
Affiliation(s)
- Jet van den Dijssel
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Veronique A. L. Konijn
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Mariël C Duurland
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Rivka de Jongh
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Lianne Koets
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- National Screening Laboratory of SanquinResearch and Laboratory ServicesAmsterdamThe Netherlands
| | - Barbera Veldhuisen
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Immunohematology DiagnosticsSanquin Diagnostic ServicesAmsterdamThe Netherlands
| | | | | | | | - Maurice Steenhuis
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Theo Rispens
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Amsterdam UMC location Vrije Universiteit AmsterdamMolecular Cell Biology and ImmunologyAmsterdamThe Netherlands
| | - C Ellen van der Schoot
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - S. Marieke van Ham
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Swammerdam Institute for Life SciencesUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Klaas P. J. M. van Gisbergen
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Physiology and Cancer Programme, Champalimaud ResearchChampalimaud FoundationLisboaPortugal
| | - Anja ten Brinke
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Carolien E. van de Sandt
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Department of Microbiology and ImmunologyUniversity of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| |
Collapse
|
37
|
Raharinirina NA, Gubela N, Börnigen D, Smith MR, Oh DY, Budt M, Mache C, Schillings C, Fuchs S, Dürrwald R, Wolff T, Hölzer M, Paraskevopoulou S, von Kleist M. SARS-CoV-2 evolution on a dynamic immune landscape. Nature 2025; 639:196-204. [PMID: 39880955 PMCID: PMC11882442 DOI: 10.1038/s41586-024-08477-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/02/2024] [Indexed: 01/31/2025]
Abstract
Since the onset of the pandemic, many SARS-CoV-2 variants have emerged, exhibiting substantial evolution in the virus' spike protein1, the main target of neutralizing antibodies2. A plausible hypothesis proposes that the virus evolves to evade antibody-mediated neutralization (vaccine- or infection-induced) to maximize its ability to infect an immunologically experienced population1,3. Because viral infection induces neutralizing antibodies, viral evolution may thus navigate on a dynamic immune landscape that is shaped by local infection history. Here we developed a comprehensive mechanistic model, incorporating deep mutational scanning data4,5, antibody pharmacokinetics and regional genomic surveillance data, to predict the variant-specific relative number of susceptible individuals over time. We show that this quantity precisely matched historical variant dynamics, predicted future variant dynamics and explained global differences in variant dynamics. Our work strongly suggests that the ongoing pandemic continues to shape variant-specific population immunity, which determines a variant's ability to transmit, thus defining variant fitness. The model can be applied to any region by utilizing local genomic surveillance data, allows contextualizing risk assessment of variants and provides information for vaccine design.
Collapse
Affiliation(s)
- N Alexia Raharinirina
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Nils Gubela
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany
- International Max-Planck Research School for Biology and Computation (IMPRS-BAC), Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | | | | | - Djin-Ye Oh
- Department 1, Robert Koch Institute, Berlin, Germany
| | - Matthias Budt
- Department 1, Robert Koch Institute, Berlin, Germany
| | | | - Claudia Schillings
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Stephan Fuchs
- Department MFI, Robert Koch Institute, Berlin, Germany
| | - Ralf Dürrwald
- Department 1, Robert Koch Institute, Berlin, Germany
| | | | - Martin Hölzer
- Department MFI, Robert Koch Institute, Berlin, Germany
| | | | - Max von Kleist
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany.
- Project Groups, Robert Koch Institute, Berlin, Germany.
| |
Collapse
|
38
|
Jantz-Naeem N, Guvencli N, Böttcher-Loschinski R, Böttcher M, Mougiakakos D, Kahlfuss S. Metabolic T-cell phenotypes: from bioenergetics to function. Am J Physiol Cell Physiol 2025; 328:C1062-C1075. [PMID: 39946684 DOI: 10.1152/ajpcell.00478.2024] [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: 07/10/2024] [Revised: 07/28/2024] [Accepted: 02/11/2025] [Indexed: 04/15/2025]
Abstract
It is well known that T-cell metabolism and function are intimately linked. Metabolic reprogramming is a dynamic process that provides the necessary energy and biosynthetic precursors while actively regulating the immune response of T cells. As such, aberrations and dysfunctions in metabolic (re)programming, resulting in altered metabolic endotypes, may have an impact on disease pathology in various contexts. With the increasing demand for personalized and highly specialized medicine and immunotherapy, understanding metabolic profiles and T-cell subset dependence on specific metabolites will be crucial to harness the therapeutic potential of immunometabolism and T cell bioenergetics. In this review, we dissect metabolic alterations in different T-cell subsets in autoimmune and viral inflammation, T cell and non-T-cell malignancies, highlighting potential anchor points for future treatment and therapeutic exploitation.
Collapse
Affiliation(s)
- Nouria Jantz-Naeem
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Nese Guvencli
- Department of Haematology, Oncology, and Cell Therapy, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Romy Böttcher-Loschinski
- Department of Haematology, Oncology, and Cell Therapy, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Martin Böttcher
- Department of Haematology, Oncology, and Cell Therapy, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Dimitrios Mougiakakos
- Department of Haematology, Oncology, and Cell Therapy, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Health and Medical Prevention, Otto-von-Guericke-University, Magdeburg, Germany
| | - Sascha Kahlfuss
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Health and Medical Prevention, Otto-von-Guericke-University, Magdeburg, Germany
- Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
39
|
Kovács Á, Hérincs Z, Papp K, Kaczmarek JZ, Larsen DN, Stage P, Bereczki L, Ujhelyi E, Pfeil T, Prechl J. In-depth immunochemical characterization of the serum antibody response using a dual-titration microspot assay. Front Immunol 2025; 16:1494624. [PMID: 40070838 PMCID: PMC11893856 DOI: 10.3389/fimmu.2025.1494624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Antigen specific humoral immunity can be characterized by the analysis of serum antibodies. While serological assays for the measurement of specific antibody levels are available, these are not quantitative in the biochemical sense. Yet, understanding humoral immune responses quantitatively on the systemic level would need a universal, complete, quantitative, comparable measurement method of antigen specific serum antibodies of selected immunoglobulin classes. Here we describe a fluorescent, dual-titration immunoassay, which provides the biochemical parameters that are both necessary and sufficient to quantitatively characterize the humoral immune response. For validation of theory, we used recombinant receptor binding domain of SARS-CoV-2 as antigen on microspot arrays and varied the concentration of both the antigen and the serum antibodies from infected persons to obtain a measurement matrix of binding data. Both titration curves were simultaneously fitted using an algorithm based on the generalized logistic function and adapted for analyzing biochemical variables of binding. We obtained equilibrium affinity constants and concentrations for distinct antibody classes. These variables reflect the quality and the effective quantity of serum antibodies, respectively. The proposed fluorescent dual-titration microspot immunoassay can generate truly quantitative serological data that is suitable for immunological, medical and systems biological analysis.
Collapse
Affiliation(s)
- Ágnes Kovács
- Department of Biostatistics, University of Veterinary Medicine Budapest, Budapest, Hungary
- Department of Applied Analysis and Computational Mathematics, Eötvös Loránd University, Budapest, Hungary
| | | | | | | | - Daniel Nyberg Larsen
- Research and Development Department, Ovodan Biotech A/S, Odense, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Pernille Stage
- Research and Development Department, Ovodan Biotech A/S, Odense, Denmark
| | | | | | - Tamás Pfeil
- Department of Applied Analysis and Computational Mathematics, Eötvös Loránd University, Budapest, Hungary
- HUN-REN–ELTE Numerical Analysis and Large Networks Research Group, Budapest, Hungary
| | | |
Collapse
|
40
|
Patel A, Lima T, Carson R, Huang Q, Bonissone SR, Castellana N. Serum proteomics reveals high-affinity and convergent antibodies by tracking SARS-CoV-2 hybrid immunity to emerging variants of concern. Front Immunol 2025; 16:1509888. [PMID: 40070844 PMCID: PMC11893383 DOI: 10.3389/fimmu.2025.1509888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/21/2025] [Indexed: 03/14/2025] Open
Abstract
The rapid spread of SARS-CoV-2 and its continuing impact on human health has prompted the need for effective and rapid development of monoclonal antibody therapeutics. In this study, we investigate polyclonal antibodies in serum and B cells from the whole blood of three donors with SARS-CoV-2 immunity to find high-affinity anti-SARS-CoV-2 antibodies to escape variants. Serum IgG antibodies were selected by their affinity to the receptor-binding domain (RBD) and non-RBD sites on the spike protein of Omicron subvariant B.1.1.529 from each donor. Antibodies were analyzed by bottom-up mass spectrometry, and matched to single- and bulk-cell sequenced repertoires for each donor. The antibodies observed in serum were recombinantly expressed, and characterized to assess domain binding, cross-reactivity between different variants, and capacity to inhibit RBD binding to host protein. Donors infected with early Omicron subvariants had serum antibodies with subnanomolar affinity to RBD that also showed binding activity to a newer Omicron subvariant BQ.1.1. The donors also showed a convergent immune response. Serum antibodies and other single- and bulk-cell sequences were similar to publicly reported anti-SARS-CoV-2 antibodies, and the characterized serum antibodies had the same variant-binding and neutralization profiles as their reported public sequences. The serum antibodies analyzed were a subset of anti-SARS-CoV-2 antibodies in the B cell repertoire, which demonstrates significant dynamics between the B cells and circulating antibodies in peripheral blood.
Collapse
Affiliation(s)
- Anand Patel
- Abterra Biosciences, Inc., San Diego, CA, United States
| | | | | | | | | | | |
Collapse
|
41
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
42
|
Vieira BM, Almeida BF, Machado MP. Eosinophil and B-cell dynamics in the milky spots from Schistosoma mansoni-infected mice: comparison with spleen and bone marrow, and extramedullary eosinopoiesis. Int Immunol 2025; 37:173-185. [PMID: 39423136 DOI: 10.1093/intimm/dxae064] [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: 09/04/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024] Open
Abstract
The milky spots (MS) are structures found in the omentum of humans and other vertebrates, representing a fraction of the lymphomyeloid tissue associated with the celom. They majorly consist of B lymphocytes, T lymphocytes, and macrophages. Also found in smaller quantities are mesothelial, stromal, dendritic, and rare mast cells. In an experimental model of Schistosoma mansoni infection, there is significant activation of the omentum and MS, which exhibit numerous eosinophils. Despite being described for many years, the complete profile of cells found in MS and their functions remains largely unexplored. Here, we evaluate the leukocyte populations of the MS in homeostasis and a murine model of S. mansoni infection. The histopathological characterization, phenotypic profile analysis, and characterization of the eosinophilic potential of progenitors and precursors comparing the MS with the spleen and bone marrow showed significant activation of MS in infected mice, with changes in the profile over the analyzed times, showing signs of migration and activation of eosinophils, with local eosinopoiesis and maintenance of the eosinophilic population. In naive mice, B1a and B1b cells make up only a small fraction of B lymphocytes. However, B1b cells expand significantly during infection, peaking at 60 days post-infection (DPI) before stabilizing by 90 DPI. B1a cells also increase initially but decrease over time. The behavior of MS differs from other primary and secondary lymphoid organs, acting as a central lymphoid organ in cavity immunity.
Collapse
Affiliation(s)
- Bruno Marques Vieira
- Laboratório de Medicina Experimental e Saúde, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Beatriz Fernandes Almeida
- Laboratório de Medicina Experimental e Saúde, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Marcelo Pelajo Machado
- Laboratório de Medicina Experimental e Saúde, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Choi M, Choi S, Cho M, Kim C. Metabolic Signaling as a Driver of T Cell Aging. Immune Netw 2025; 25:e14. [PMID: 40078788 PMCID: PMC11896665 DOI: 10.4110/in.2025.25.e14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Aging significantly diminishes T cell immunity, increasing susceptibility to infections and reducing vaccine efficacy in older individuals. Metabolism plays a key role in T cell function, shaping their energy requirements, activation, and differentiation. Recent studies highlight altered metabolic signaling as a pivotal factor in T cell aging, influencing the ability of T cells to maintain quiescence, respond to activation, and differentiate into functional subsets. Aberrant metabolic pathways disrupt the quiescence of aged T cells and skew their differentiation toward short-lived, pro-inflammatory effector T cells while hindering the generation of long-lived memory and T follicular helper cells. These changes contribute to a hyper-inflammatory state, exacerbate chronic low-grade inflammation, and compromise immune homeostasis. In this review, we explore how metabolic signaling is altered during T cell aging and the resulting functional impacts. We also discuss therapeutic approaches aimed at restoring proper T cell differentiation, improving vaccine responses, and rejuvenating immune function in older populations.
Collapse
Affiliation(s)
- Minju Choi
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02708, Korea
| | - Sujin Choi
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02708, Korea
| | - Minkyeong Cho
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02708, Korea
| | - Chulwoo Kim
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02708, Korea
| |
Collapse
|
44
|
Ceraolo MG, Leccese M, Cassotta A, Triolo S, Bombaci M, Coluccio E, Prati D, Ungaro R, Abrignani S, Bandera A, Sallusto F, Lanzavecchia A, Notarbartolo S. Dual Activation-Induced Marker Combinations Efficiently Identify and Discern Antigen-Specific and Bystander-Activated Human CD4 + T Cells. Eur J Immunol 2025; 55:e202451404. [PMID: 39663678 PMCID: PMC11830384 DOI: 10.1002/eji.202451404] [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: 07/22/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
Identifying activated T lymphocytes and differentiating antigen-specific from bystander T cells is crucial for understanding adaptive immune responses. This study investigates the efficacy of activation-induced markers (AIMs) in distinguishing these cell populations. We measured the expression of commonly used AIMs (CD25, CD38, CD40L, CD69, CD137, HLA-DR, ICOS, and OX40) in an in vitro T-cell activation system and evaluated their sensitivity, specificity, and positive predictive value. We demonstrated that individual AIMs, while specific in detecting activated CD4+ T cells, poorly discriminate between antigen-specific and bystander activation, as assessed by a discriminative capacity (DC) score we developed. Our analysis revealed that dual AIM combinations significantly enhanced the ability to distinguish antigen-specific from bystander-activated T cells, achieving DC scores above 90%. These combinations also improved positive predictive value and specificity with a modest reduction in sensitivity. The CD25hi/ICOShi combination emerged as the most efficient, with an average sensitivity of 84.35%, specificity of 99.7%, and DC score of 90.12%. Validation through T-cell cloning and antigen re-stimulation confirmed the robustness of our predictions. This study provides a practical framework for researchers to optimize strategies for identifying and isolating antigen-specific human CD4+ T lymphocytes and studying their phenotype, function, and T-cell receptor repertoire.
Collapse
Affiliation(s)
- Maria Grazia Ceraolo
- INGM, Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
| | - Maristella Leccese
- INGM, Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
| | - Antonino Cassotta
- Institute for Research in BiomedicineUniversità della Svizzera italianaBellinzonaSwitzerland
| | - Sara Triolo
- INGM, Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
| | - Mauro Bombaci
- INGM, Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
| | - Elena Coluccio
- Department of Transfusion Medicine and HematologyFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Daniele Prati
- Department of Transfusion Medicine and HematologyFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Riccardo Ungaro
- Infectious Diseases UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Sergio Abrignani
- INGM, Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Alessandra Bandera
- Infectious Diseases UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
- Centre for Multidisciplinary Research in Health Science (MACH)Università degli Studi di MilanoMilanItaly
| | - Federica Sallusto
- Institute for Research in BiomedicineUniversità della Svizzera italianaBellinzonaSwitzerland
- Institute of Microbiology, ETH ZurichZurichSwitzerland
| | - Antonio Lanzavecchia
- INGM, Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
| | - Samuele Notarbartolo
- Infectious Diseases UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
45
|
Zhao Y, He C, Peng M, Li M, Liu X, Han X, Fu Q, Wu Y, Yue F, Yan C, Zhao G, Shen C. Large-Scale Screening of CD4 + T-Cell Epitopes From SARS-CoV-2 Proteins and the Universal Detection of SARS-CoV-2 Specific T Cells for Northeast Asian Population. J Med Virol 2025; 97:e70241. [PMID: 39977358 DOI: 10.1002/jmv.70241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/09/2024] [Accepted: 01/25/2025] [Indexed: 02/22/2025]
Abstract
The polymorphism of human leukocyte antigens in the Northeast Asian populations and the lack of broad-spectrum T-cell epitopes covering this cohort markedly limited the development of T cell-directed vaccines against SARS-CoV-2 infection, and also hampered the universal detection of SARS-CoV-2 specific T cells. In this study, 93 CD4+ T-cell epitopes restricted by 12 prevalent HLA-DRB1 allotypes, which covering over 80% Chinese and Northeast Asian populations, were identified from the S, E, M, N and RdRp proteins of SARS-CoV-2 by in silico prediction, DC-peptide-PBL coculture experiment, and immunization in HLA-A2/DR1 transgenic mice. Furthermore, by using validated 215 CD8+ T cell epitope peptides and 123 CD4+ T-cell epitope peptides covering Northeast Asian cohort, the universal ELISpot detection systems of SARS-CoV-2 specific CD8+ T cells and CD4+ T cells were established, for the first time, and followed by the tests for 50 unexposed and 100 convalescent samples. The median of spot-forming units for CD8+ T cells and CD4+ T cells were 68 and 15, respectively, in the unexposed donors, but were 137 and 52 in the convalescent donors 6 months after recovery while 128 and 47 in the convalescent donors 18 months after recovery. This work initially provided the broad-spectrum CD4+ T-cell epitope library of SARS-CoV-2 for the design of T cell-directed vaccines and the universal T cell detection tool tailoring to Northeast Asian population, and confirmed the long-term memory T cell immunity after SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Chengtao He
- Nanjing Red Cross Blood Center, Nanjing, China
| | - Min Peng
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Min Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Xiaotao Liu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Xuelian Han
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Qiang Fu
- Nanjing Red Cross Blood Center, Nanjing, China
| | - Yandan Wu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Fangping Yue
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Chunguang Yan
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Chuanlai Shen
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| |
Collapse
|
46
|
Maison DP, Tasissa H, Deitchman A, Peluso MJ, Deng Y, Miller FD, Henrich TJ, Gerschenson M. COVID-19 clinical presentation, management, and epidemiology: a concise compendium. Front Public Health 2025; 13:1498445. [PMID: 39957982 PMCID: PMC11826932 DOI: 10.3389/fpubh.2025.1498445] [Citation(s) in RCA: 1] [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: 09/27/2024] [Accepted: 01/21/2025] [Indexed: 02/18/2025] Open
Abstract
Coronavirus Disease 2019, caused by severe acute respiratory coronavirus 2, has been an ever-evolving disease and pandemic, profoundly impacting clinical care, drug treatments, and understanding. In response to this global health crisis, there has been an unprecedented increase in research exploring new and repurposed drugs and advancing available clinical interventions and treatments. Given the widespread interest in this topic, this review aims to provide a current summary-for interested professionals not specializing in COVID-19-of the clinical characteristics, recommended treatments, vaccines, prevention strategies, and epidemiology of COVID-19. The review also offers a historical perspective on the pandemic to enhance understanding.
Collapse
Affiliation(s)
- David P. Maison
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Hawi Tasissa
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Amelia Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Michael J. Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - F. DeWolfe Miller
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Timothy J. Henrich
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| |
Collapse
|
47
|
Hromić-Jahjefendić A, Aljabali AAA. Analysis of the immune response in COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:31-71. [PMID: 40246347 DOI: 10.1016/bs.pmbts.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The COVID-19 pandemic, instigated by the novel coronavirus SARS-CoV-2, has emerged as a significant global health challenge, demanding a profound grasp of the immune response. The innate immune system, a multifaceted network encompassing pattern recognition receptors (PRRs) and effector cells, assumes a pivotal function in detecting and countering this viral assailant. Toll-like receptors (TLRs), situated on immune cell surfaces and within endosomes, play a central role in recognizing SARS-CoV-2. TLR-2 and TLR-4 discern specific viral constituents, such as the spike (S) protein, setting off inflammatory signaling cascades and catalyzing the generation of type I interferons. Intracellular PRRs, including the RIG-I-like receptors (RLRs), RIG-I and MDA5, detect viral RNA within the cytoplasm of infected cells, provoking antiviral responses by initiating the synthesis of type I interferons. The equilibrium between interferons and pro-inflammatory cytokines dictates the outcomes of the disease. Interferons play an indispensable role in governing viral replication, while unregulated cytokine production can result in tissue harm and inflammation. This intricate dynamic underpins therapeutic strategies aimed at regulating immune responses in individuals grappling with COVID-19. Natural killer (NK) cells, with their capacity to recognize infected cells through the "missing self" phenomenon and activating receptors, make significant contributions to the defense against SARS-CoV-2. NK cells play a pivotal role in eliminating infected cells and boosting immune responses through antibody-dependent cell-mediated cytotoxicity (ADCC). In conclusion, comprehending the interplay among PRRs, interferons, and NK cells within innate immunity is paramount for discerning and combatting SARS-CoV-2. This comprehension illuminates therapeutic interventions and vaccine development, casting light on our endeavors to confront this worldwide health crisis.
Collapse
Affiliation(s)
- Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina.
| | - Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| |
Collapse
|
48
|
Autaa G, Papagno L, Nogimori T, Boizard-Moracchini A, Korenkov D, Roy M, Suzuki K, Masuta Y, White E, Llewellyn-Lacey S, Yoshioka Y, Nicoli F, Price DA, Dechanet-Merville J, Yamamoto T, Pellegrin I, Appay V. Aging and inflammation limit the induction of SARS-CoV-2-specific CD8+ T cell responses in severe COVID-19. JCI Insight 2025; 10:e180867. [PMID: 39847442 PMCID: PMC11949069 DOI: 10.1172/jci.insight.180867] [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: 03/04/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
CD8+ T cells are critical for immune protection against severe COVID-19 during acute infection with SARS-CoV-2. However, the induction of antiviral CD8+ T cell responses varies substantially among infected people, and a better understanding of the mechanisms that underlie such immune heterogeneity is required for pandemic preparedness and risk stratification. In this study, we analyzed SARS-CoV-2-specific CD4+ and CD8+ T cell responses in relation to age, clinical status, and inflammation among patients infected primarily during the initial wave of the pandemic in France or Japan. We found that age-related contraction of the naive lymphocyte pool and systemic inflammation were associated with suboptimal SARS-CoV-2-specific CD4+ and, even more evidently, CD8+ T cell immunity in patients with acute COVID-19. No such differences were observed for humoral immune responses targeting the spike protein of SARS-CoV-2. We also found that the proinflammatory cytokine IL-18, concentrations of which were significantly elevated among patients with severe disease, suppressed the de novo induction and memory recall of antigen-specific CD8+ T cells, including those directed against SARS-CoV-2. These results potentially explain the vulnerability of older adults to infections that elicit a profound inflammatory response, exemplified by acute COVID-19.
Collapse
Affiliation(s)
- Gaëlle Autaa
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
| | - Laura Papagno
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
| | - Takuto Nogimori
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | | | - Daniil Korenkov
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
| | - Maeva Roy
- CHU Bordeaux, Laboratory of Immunology and Immunogenetics, 33000 Bordeaux, France
| | - Koichiro Suzuki
- The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Osaka, Japan
| | - Yuji Masuta
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Eoghann White
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
| | - Sian Llewellyn-Lacey
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Yasuo Yoshioka
- The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Osaka, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, and
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Francesco Nicoli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - David A. Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Julie Dechanet-Merville
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
| | - Takuya Yamamoto
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Isabelle Pellegrin
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
- CHU Bordeaux, Laboratory of Immunology and Immunogenetics, 33000 Bordeaux, France
| | - Victor Appay
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| |
Collapse
|
49
|
Guironnet-Paquet A, Hamzeh-Cognasse H, Berard F, Cognasse F, Richard JC, Yonis H, Mezidi M, Desebbe O, Delannoy B, Demeret S, Marois C, Saheb S, Le QV, Schoeffler M, Pugliesi PS, Debord S, Bastard P, Cobat A, Casanova JL, Pescarmona R, Viel S, Nicolas JF, Nosbaum A, Vocanson M, Hequet O. Therapeutic plasma exchange accelerates immune cell recovery in severe COVID-19. Front Immunol 2025; 15:1492672. [PMID: 39896810 PMCID: PMC11782122 DOI: 10.3389/fimmu.2024.1492672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/04/2024] [Indexed: 02/04/2025] Open
Abstract
Background Immunological disturbances (anti-type I IFN auto-antibody production, cytokine storm, lymphopenia, T-cell hyperactivation and exhaustion) are responsible for disease exacerbation during severe COVID-19 infections. Methods In this study, we set up a prospective, randomised clinical trial (ClinicalTrials.gov ID: NCT04751643) and performed therapeutic plasma exchange (TPE) in severe COVID-19 patients in order to decrease excess cytokines and auto-antibodies and to assess whether adding TPE to the standard treatment (ST, including corticosteroids plus high-flow rate oxygen) could help restore immune parameters and limit the progression of acute respiratory distress syndrome (ARDS). Results As expected, performing TPE decreased the amount of anti-type I IFN auto-antibodies and improved the elimination or limited the production of certain inflammatory mediators (IL-18, IL-7, CCL2, CCL3, etc.) circulating in the blood of COVID-19 patients, compared to ST controls. Interestingly, while TPE did not influence changes in ARDS parameters throughout the protocol, it proved more effective than ST in reversing lymphopenia, preventing T-cell hyperactivation and reducing T-cell exhaustion, notably in a fraction of TPE patients who had an early favourable respiratory outcome. TPE also restored appropriate numbers of CD4+ and CD8+ T-cell memory populations and increased the number of circulating virus-specific T cells in these patients. Conclusion Our results therefore indicate that the addition of TPE sessions to the standard treatment accelerates immune cell recovery and contributes to the development of appropriate antiviral T-cell responses in some patients with severe COVID-19 disease.
Collapse
Affiliation(s)
- Aurelie Guironnet-Paquet
- Apheresis Unit, Etablissement Français du Sang Auvergne-Rhône-Alpes, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre Bénite, France
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| | - Hind Hamzeh-Cognasse
- University of Jean Monnet, Mines Saint-Étienne, Institut National de la Santé et de la Recherche Médicale (INSERM), U 1059 SAINBIOSE, Saint-Étienne, France
| | - Frederic Berard
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Fabrice Cognasse
- University of Jean Monnet, Mines Saint-Étienne, Institut National de la Santé et de la Recherche Médicale (INSERM), U 1059 SAINBIOSE, Saint-Étienne, France
- Scientific Department, Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France
| | - Jean Christophe Richard
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Hodane Yonis
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Mehdi Mezidi
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Olivier Desebbe
- Department of Anesthesiology and Perioperative Medicine, Sauvegarde Clinic, Ramsay Santé, Lyon, France
| | - Bertrand Delannoy
- Department of Anesthesiology and Perioperative Medicine, Sauvegarde Clinic, Ramsay Santé, Lyon, France
| | - Sophie Demeret
- Neuro-Intensive Care Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Clemence Marois
- Neuro-Intensive Care Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, Institut du Cerveau et de la Moelle (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Departement Médico-Universitaire (DMU) Neurosciences 6, Paris, France
- Groupe de Recherche Clinique en REanimation et Soins Intensifs du Patient en Insuffisance Respiratoire aiguE (GRC-RESPIRE), Sorbonne Université, Paris, France
| | - Samir Saheb
- Hemobiotherapy Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Quoc Viet Le
- Intensive Care Unit, Medipôle Lyon Villeurbanne, Villeurbanne, France
| | - Mathieu Schoeffler
- Department of Anesthesiology and Intensive Care Unit, Centre Hospitalier de Montélimar, Montélimar, France
| | - Paul Simon Pugliesi
- Intensive Care Unit, Centre Hospitalier William Morey, Chalon sur Saône, France
| | - Sophie Debord
- Department of Anesthesiology and Intensive Care Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon (HCL), Lyon, France
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique des Hopitaux de Paris (AP-HP), Paris, France
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
| | - Jean Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique des Hopitaux de Paris (AP-HP), Paris, France
- Howards Hugues Medical Institute, New York, NY, United States
| | - Rémi Pescarmona
- Immun Monitorage Laboratory, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Sébastien Viel
- Plateforme de Biothérapies et de production de Médicaments de Thérapie Innovante (MTI), Hôpital Edouard Herriot, Hospices Civils de Lyon (HCL), Lyon, France
| | - Jean François Nicolas
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Audrey Nosbaum
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Marc Vocanson
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| | - Olivier Hequet
- Apheresis Unit, Etablissement Français du Sang Auvergne-Rhône-Alpes, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre Bénite, France
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| |
Collapse
|
50
|
Avadhanula V, Creighton CJ, Ferlic-Stark L, Nagaraj D, Zhang Y, Sucgang R, Nicholson EG, Rajan A, Menon VK, Doddapaneni H, Muzny DM, Metcalf GA, Cregeen SJJ, Hoffman KL, Gibbs RA, Petrosino JF, Piedra PA. Longitudinal host transcriptional responses to SARS-CoV-2 infection in adults with extremely high viral load. PLoS One 2025; 20:e0317033. [PMID: 39820858 PMCID: PMC11737797 DOI: 10.1371/journal.pone.0317033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025] Open
Abstract
Current understanding of viral dynamics of SARS-CoV-2 and host responses driving the pathogenic mechanisms in COVID-19 is rapidly evolving. Here, we conducted a longitudinal study to investigate gene expression patterns during acute SARS-CoV-2 illness. Cases included SARS-CoV-2 infected individuals with extremely high viral loads early in their illness, individuals having low SARS-CoV-2 viral loads early in their infection, and individuals testing negative for SARS-CoV-2. We could identify widespread transcriptional host responses to SARS-CoV-2 infection that were initially most strongly manifested in patients with extremely high initial viral loads, then attenuating within the patient over time as viral loads decreased. Genes correlated with SARS-CoV-2 viral load over time were similarly differentially expressed across independent datasets of SARS-CoV-2 infected lung and upper airway cells, from both in vitro systems and patient samples. We also generated expression data on the human nose organoid model during SARS-CoV-2 infection. The human nose organoid-generated host transcriptional response captured many aspects of responses observed in the above patient samples, while suggesting the existence of distinct host responses to SARS-CoV-2 depending on the cellular context, involving both epithelial and cellular immune responses. Our findings provide a catalog of SARS-CoV-2 host response genes changing over time and magnitude of these host responses were significantly correlated to viral load.
Collapse
Affiliation(s)
- Vasanthi Avadhanula
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Chad J. Creighton
- Dan L. Duncan Comprehensive Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX, United States of America
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Laura Ferlic-Stark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Divya Nagaraj
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX, United States of America
| | - Richard Sucgang
- Houston Methodist Research Institute, Center for Health Data Science and Analytics, Houston, Texas, United States of America
| | - Erin G. Nicholson
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Anubama Rajan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Vipin Kumar Menon
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Harshavardhan Doddapaneni
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Donna Marie Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ginger A. Metcalf
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sara Joan Javornik Cregeen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Kristi Louise Hoffman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Joseph F. Petrosino
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Pedro A. Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|