1
|
Tripathy AS, Singh D, Trimbake D, Salwe S, Tripathy S, Kakrani A, Jali P, Chavan H, Yadav P, Sahay R, Sarje P, Babar P, Shete A, Nandapurkar A, Kulkarni M. Humoral and cellular immune response to AZD1222 /Covishield and BV152/Covaxin COVID-19 vaccines among adults in India. Hum Vaccin Immunother 2024; 20:2410579. [PMID: 39434214 PMCID: PMC11497953 DOI: 10.1080/21645515.2024.2410579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/06/2024] [Accepted: 09/25/2024] [Indexed: 10/23/2024] Open
Abstract
Several COVID-19 vaccines were developed using different approaches to prevent both symptomatic COVID-19 cases and fatalities. The adults were vaccinated with two doses of AZD1222/Covishield (n = 77) [manufactured by Serum Institute of India Pvt Ltd] vaccine and BV152/Covaxin (n = 99) [manufactured by Bharat Biotech] vaccine. They were assessed for immune response at pre-vaccination, 1 month post first and 6 months post second dose for anti-SARS-CoV-2 IgG antibody, surrogate neutralizing antibody (NAbs), immune phenotypes, antigen specific NK, B and T cell response, their effector functionality by ELISPOT and plasma cytokine profile. Both vaccines elicited enhanced IgG antibody and Nab levels compared to the baseline. BV152/Covaxin, the whole virus inactivated vaccine exhibited higher IgG (70% vs 100%), Nab (90% vs 100%), and robust T cell (31% vs 96%) responses at 6 months post second dose compared to 1 month post first dose justifying the utility of the second dose. Detection of SARS-CoV-2 WV and S1 specific CD4+ central T cell memory response in AZD1222/Covishield vaccinee at 6 months post second dose and higher CD4+ and CD8+ naïve and central memory T cell response in BV152/Covaxin vaccinee at 1 month post first dose indicated the involvement of memory T cells. Persistent IgG and NAb responses along with IgG+B and IgG+memory B cells in AZD1222/Covishield recipients at 6 months post second dose indicated sustained immune memory response. Continued heightened IFN-γ secreting T cell response (ELISPOT) displayed by both the vaccine platforms could serve as a co correlate of protection, further to evaluation in follow up studies. Overall, our data suggest that coordinated functions of humoral and cellular branches of adaptive immunity may pave ways toward protective immunity against COVID-19.
Collapse
Affiliation(s)
| | | | | | | | - Srikanth Tripathy
- Dr. D. Y. Patil Medical College, Hospital & Research Centre, Pune, India
| | - Arjun Kakrani
- Dr. D. Y. Patil Medical College, Hospital & Research Centre, Pune, India
| | - Priyanka Jali
- Dr. D. Y. Patil Medical College, Hospital & Research Centre, Pune, India
| | - Hanmant Chavan
- Dr. D. Y. Patil Medical College, Hospital & Research Centre, Pune, India
| | - Pragya Yadav
- ICMR-National Institute of Virology, Pune, India
| | - Rima Sahay
- ICMR-National Institute of Virology, Pune, India
| | | | - Prasad Babar
- ICMR-National Institute of Virology, Pune, India
| | - Anita Shete
- ICMR-National Institute of Virology, Pune, India
| | | | | |
Collapse
|
2
|
Blasco A, Royuela A, García-Gómez S, Gómez-Lozano N, Sánchez-Arjona A, de la Fuente J, Anel J, Sánchez-Galarraga I, Pérez-Redondo M, González E, Silva L. Association of SARS-CoV-2 immunoserology and vaccination status with myocardial infarction severity and outcome. Vaccine 2024; 42:126305. [PMID: 39244425 DOI: 10.1016/j.vaccine.2024.126305] [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: 05/04/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND The COVID-19 pandemic adversely affected the severity and prognosis of patients with acute myocardial infarction (MI) caused by atherothrombosis (type 1 MI). The effect, if any, of COVID-19 vaccination and natural SARS-CoV2 serologic immunity in these patients is unclear. Our aim was to analyze the association between the severity and outcome of patients with type 1 MI and their previous SARS-CoV2 vaccination and serostatus. METHODS A single-center retrospective cohort study conducted between March 1, 2020 and March 1, 2023. Clinical and follow-up information was collected from medical records and patients. Total antibodies (IgM, IgA, IgG) to nucleocapsid (N) antigens were measured by ECLIA (electrochemiluminescence-based immunoassay) to test the immune response to natural infection. If positive, IgM and IgG antibodies to spike (S) surface antigens were measured by CLIA to test the immune response to vaccine or natural infection. Multivariable logistic regression analysis was performed, adjusting for age, sex, hypertension, diabetes, and dyslipidemia. RESULTS Total sample of 949 patients, 656 with ST-segment elevation MI (STEMI) and 293 with non-ST-segment elevation MI (NSTEMI). Mean age was 64 (SD 13) years, 80 % men. Pre-admission vaccination status was: ≥ 1 dose, 53 % of patients; complete vaccination, 49 %; first booster dose, 25 %. The majority (84 %) of vaccines administered were mRNA-based. Six months after MI, 92 (9.7 %) patients had a major adverse cardiac event (MACE) and 50 died; 11 % of patients had severe heart failure or cardiogenic shock (Killip III-IV) after STEMI. Vaccinated patients with STEMI and positive serology (Pos/Vax group) had a higher risk of Killip III-IV on admission: OR 2.63 (1.27-5.44), p = 0.010. SARS-CoV-2 S-specific IgG titers were highest in this group (median > 2080 AU/mL, [IQR 1560- >2080] vs 91 [32-198] in the unvaccinated group). In the overall sample, a higher incidence of 6-month MACE was not demonstrated (OR 1.89 [0.98-3.61], p = 0.055). CONCLUSIONS The combination of vaccination and natural SARS-CoV2 infection was associated with the development of severe heart failure and cardiogenic shock in patients with STEMI, possibly related to an increased serological response.
Collapse
Affiliation(s)
- Ana Blasco
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain; Research Ethics Committee, Instituto de Investigación Puerta de Hierro-Segovia de Arana, Madrid, Spain.
| | - Ana Royuela
- Biostatistics Unit, Instituto de Investigación Puerta de Hierro-Segovia de Arana, Madrid, Spain; Center for Biomedical Research in Epidemiology and Public Health Network (CIBERESP), Madrid, Spain
| | - Sergio García-Gómez
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Natalia Gómez-Lozano
- Immunology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Alberto Sánchez-Arjona
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Jorge de la Fuente
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Jorge Anel
- Microbiology Department, Serology Section, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | | | - Marina Pérez-Redondo
- Intensive Care Unit, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Elisa González
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Lorenzo Silva
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| |
Collapse
|
3
|
Megha KB, Reshma S, Amir S, Krishnan MJA, Shimona A, Alka R, Mohanan PV. Comprehensive Risk Assessment of Infection Induced by SARS-CoV-2. Mol Neurobiol 2024; 61:9851-9872. [PMID: 37817031 DOI: 10.1007/s12035-023-03682-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023]
Abstract
The pandemic COVID-19 (coronavirus disease 2019) is caused by the severe acute respiratory syndrome corona virus 2 (SARS-CoV-2), which devastated the global economy and healthcare system. The infection caused an unforeseen rise in COVID-19 patients and increased the mortality rate globally. This study gives an overall idea about host-pathogen interaction, immune responses to COVID-19, recovery status of infection, targeted organs and complications associated, and comparison of post-infection immunity in convalescent subjects and non-infected individuals. The emergence of the variants and episodes of COVID-19 infections made the situation worsen. The timely introduction of vaccines and precautionary measures helped control the infection's severity. Later, the population that recovered from COVID-19 grew significantly. However, understanding the impact of healthcare issues resulting after infection is paramount for improving an individual's health status. It is now recognised that COVID-19 infection affects multiple organs and exhibits a broad range of clinical manifestations. So, post COVID-19 infection creates a high risk in individuals with already prevailing health complications. The identification of post-COVID-19-related health issues and their appropriate management is of greater importance to improving patient's quality of life. The persistence, sequelae and other medical complications that normally last from weeks to months after the recovery of the initial infection are involved with COVID-19. A multi-disciplinary approach is necessary for the development of preventive measures, techniques for rehabilitation and strategies for clinical management when it comes to long-term care.
Collapse
Affiliation(s)
- K B Megha
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - S Reshma
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - S Amir
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - M J Ajai Krishnan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - A Shimona
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - Rao Alka
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - P V Mohanan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India.
| |
Collapse
|
4
|
Li K, Wu Y, Zhang H, Chen S, Wu B, Li T, Li E, Luo F, Jin A, Zhang B, Zhang Y, Gong R, Zhang H, Chiu S. A Novel Circular Delta-XBB15 RBD Dimeric Protein Subunit Vaccine Mediated by Split Intein Elicits an Immune Response and Protection Against Multiple SARS-CoV-2 Variants in Mice. J Med Virol 2024; 96:e70134. [PMID: 39711477 DOI: 10.1002/jmv.70134] [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: 08/05/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/24/2024]
Abstract
SARS-CoV-2 continues to mutate, leading to breakthrough infections. The development of new vaccine strategies to combat various strains is crucial. Protein cyclization can enhance thermal stability and may improve immunogenicity. Here, we designed a cyclic tandem dimeric receptor-binding domain protein (cirRBD2) via the split intein Cth-Ter. Cyclization does not affect the antigen epitopes of RBD but results in better thermal stability than that of its linear counterpart (linRBD2). Compared with the mice immunized with linRBD2, those immunized with two doses of 5 μg of cirRBD2 produced significantly greater levels of broad-spectrum neutralizing antibodies, and generated a considerable cellular immune response. In the VEEV-VRP-hACE2-transduced mouse model, two doses of 5 μg of cirRBD2 provided protection against infection with BA.5, XBB.1.9, and partial protection against EG.5 which has more mutations. This study developed a novel circular RBD dimer subunit vaccine for SARS-CoV-2 that exhibits broad-spectrum neutralizing activity against various variants. A similar strategy can be applied to develop vaccines for other pathogens, especially for thermally stable vaccines.
Collapse
MESH Headings
- Animals
- Mice
- COVID-19 Vaccines/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- COVID-19/prevention & control
- COVID-19/immunology
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/genetics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Inteins
- Mice, Inbred BALB C
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/chemistry
- Humans
- Female
- Immunity, Cellular
Collapse
Affiliation(s)
- Kangyin Li
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Yan Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Hongqing Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Shaohong Chen
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Bihao Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Tingting Li
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Feiyang Luo
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing, China
| | - Aishun Jin
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing, China
| | - Bo Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yanan Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Rui Gong
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Huajun Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| |
Collapse
|
5
|
Wang L, Xu R, Huang D, Peng P, Sun K, Hu J, Liu BZ, Fang L, Zhang L, Sun X, Gu F, Tang N, Huang AL, Lin X, Lan X. Identification of virus epitopes and reactive T-cell receptors from memory T cells without peptide synthesis. Commun Biol 2024; 7:1432. [PMID: 39496850 PMCID: PMC11535475 DOI: 10.1038/s42003-024-07048-x] [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: 06/11/2024] [Accepted: 10/10/2024] [Indexed: 11/06/2024] Open
Abstract
Identifying epitopes and their corresponding T-cell receptor (TCR) sequences is crucial in the face of rapidly mutating viruses. Peptide synthesis is often required to confirm the exact epitope sequences, which is time-consuming and expensive. In this study, we introduce a scalable workflow to identify the exact sequences of virus epitopes and reactive TCRs targeting the epitopes from memory T cells. Following the narrowing down of epitopes to specific regions via the tandem minigene (TMG) system, our workflow incorporates the utilization of peptide-major histocompatibility complex-displaying yeasts (pMHC-displaying yeasts) to rapidly screen immunogenic epitopes' precise sequences, obviating the necessity for the chemical synthesis of peptides. Focusing on SARS-CoV-2, we identify the precise sequences of reactive TCRs, targeting conserved epitopes across the Coronaviridae family, from the blood of COVID-19-recovered individuals over 8 months. Notably, we reveal that at least 75% (6/8) of the tested donors harbor T cells targeting a shared epitope, KTFPPTEPK, derived from the N protein. Furthermore, several identified TCRs exhibit cross-reactivity to mutant epitopes, suggesting a potential mechanism for sustained T-cell responses against emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Lihui Wang
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Runda Xu
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Daosheng Huang
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Pai Peng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Keyong Sun
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Jie Hu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Bei-Zhong Liu
- Yong-Chuan Hospital of Chongqing Medical University, Chongqing, China
| | - Liang Fang
- Yong-Chuan Hospital of Chongqing Medical University, Chongqing, China
| | - Liwen Zhang
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Xin Sun
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Fei Gu
- Alibaba Group, 311121, Hangzhou, China.
| | - Ni Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
| | - Ai-Long Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
| | - Xin Lin
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China.
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.
| | - Xun Lan
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China.
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
6
|
Deng S, Xu Z, Hu J, Yang Y, Zhu F, Liu Z, Zhang H, Wu S, Jin T. The molecular mechanisms of CD8 + T cell responses to SARS-CoV-2 infection mediated by TCR-pMHC interactions. Front Immunol 2024; 15:1468456. [PMID: 39450171 PMCID: PMC11499136 DOI: 10.3389/fimmu.2024.1468456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024] Open
Abstract
Cytotoxic CD8+ T lymphocytes (CTLs) have been implicated in the severity of COVID-19. The TCR-pMHC ternary complex, formed by the T cell receptor (TCR) and peptide-MHC (major histocompatibility complex), constitutes the molecular basis of CTL responses against SARS-CoV-2. While numerous studies have been conducted on T cell immunity, the molecular mechanisms underlying CTL-mediated immunity against SARS-CoV-2 infection have not been well elaborated. In this review, we described the association between HLA variants and different immune responses to SARS-CoV-2 infection, which may lead to varying COVID-19 outcomes. We also summarized the specific TCR repertoires triggered by certain SARS-CoV-2 CTL epitopes, which might explain the variations in disease outcomes among different patients. Importantly, we have highlighted the primary strategies used by SARS-CoV-2 variants to evade T-cell killing: disrupting peptide-MHC binding, TCR recognition, and antigen processing. This review provides valuable insights into the molecule mechanism of CTL responses during SARS-CoV-2 infection, aiding efforts to control the pandemic and prepare for future challenges.
Collapse
Affiliation(s)
- Shasha Deng
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhihao Xu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jing Hu
- Laboratory of Structural Immunology, the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yunru Yang
- Laboratory of Structural Immunology, the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fang Zhu
- Laboratory of Structural Immunology, the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhuan Liu
- Laboratory of Structural Immunology, the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongliang Zhang
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Songquan Wu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Tengchuan Jin
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Laboratory of Structural Immunology, the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science & Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| |
Collapse
|
7
|
Tsagkli P, Geropeppa M, Papadatou I, Spoulou V. Hybrid Immunity against SARS-CoV-2 Variants: A Narrative Review of the Literature. Vaccines (Basel) 2024; 12:1051. [PMID: 39340081 PMCID: PMC11436074 DOI: 10.3390/vaccines12091051] [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: 08/11/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
The emergence of SARS-CoV-2 led to a global health crisis and the burden of the disease continues to persist. The rapid development and emergency authorization of various vaccines, including mRNA-based vaccines, played a pivotal role in mitigating severe illness and mortality. However, rapid viral mutations, leading to several variants of concern, challenged vaccine effectiveness, particularly concerning immune evasion. Research on immunity, both from natural infection and vaccination, revealed that while neutralizing antibodies provide protection against infection, their effect is short-lived. The primary defense against severe COVID-19 is derived from the cellular immune response. Hybrid immunity, developed from a combination of natural infection and vaccination, offers enhanced protection, with convalescent vaccinated individuals showing significantly higher levels of neutralizing antibodies. As SARS-CoV-2 continues to evolve, understanding the durability and breadth of hybrid immunity becomes crucial. This narrative review examines the latest data on humoral and cellular immunity from both natural infection and vaccination, discussing how hybrid immunity could inform and optimize future vaccination strategies in the ongoing battle against COVID-19 and in fear of a new pandemic.
Collapse
Affiliation(s)
- Panagiota Tsagkli
- Immunobiology and Vaccinology Research Laboratory and Infectious Diseases Department "MAKKA", First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Athens Medical School, 11527 Athens, Greece
| | - Maria Geropeppa
- Immunobiology and Vaccinology Research Laboratory and Infectious Diseases Department "MAKKA", First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Athens Medical School, 11527 Athens, Greece
| | - Ioanna Papadatou
- Immunobiology and Vaccinology Research Laboratory and Infectious Diseases Department "MAKKA", First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Athens Medical School, 11527 Athens, Greece
| | - Vana Spoulou
- Immunobiology and Vaccinology Research Laboratory and Infectious Diseases Department "MAKKA", First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Athens Medical School, 11527 Athens, Greece
| |
Collapse
|
8
|
McMahon WC, Kwatra G, Izu A, Jones SA, Mbele NJ, Jafta N, Lala R, Shalekoff S, Tiemessen CT, Madhi SA, Nunes MC. T-cell responses to ancestral SARS-CoV-2 and Omicron variant among unvaccinated pregnant and postpartum women living with and without HIV in South Africa. Sci Rep 2024; 14:20348. [PMID: 39223211 PMCID: PMC11369237 DOI: 10.1038/s41598-024-70725-8] [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: 12/18/2023] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
SARS-CoV-2 cell-mediated immunity remains understudied during pregnancy in unvaccinated Black African women living with HIV (WLWH) from low- and middle-income countries. We investigated SARS-CoV-2-specific T-cell responses 1 month following infection in 24 HIV-uninfected women and 15 WLWH at any stage during pregnancy or postpartum. The full-length spike (FLS) glycoprotein and nucleocapsid (N) protein of wild-type (WT) SARS-CoV-2, as well as mutated spike protein regions found in the Omicron variant (B.1.1.529) were targeted by flow cytometry. WT-specific CD4+ and CD8+ T cells elicited similar FLS- and N-specific responses in HIV-uninfected women and WLWH. SARS-CoV-2-specific T-lymphocytes were predominantly TNF-α monofunctional in pregnant and postpartum women living with and without HIV, with fever cells producing either IFN-γ or IL-2. Furthermore, T-cell responses were unaffected by Omicron-specific spike mutations as similar responses between Omicron and the ancestral virus were detected for CD4+ and CD8+ T cells. Our results collectively demonstrate comparable T-cell responses between WLWH on antiretroviral therapy and HIV-uninfected pregnant and postpartum women who were naïve to Covid-19 vaccination. Additionally, we show that T cells from women infected with the ancestral virus, Beta variant (B.1.351), or Delta variant (B.1.617.2) can cross-recognize Omicron, suggesting an overall preservation of T-cell immunity.
Collapse
Affiliation(s)
- William C McMahon
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Research Chair Initiative in Vaccine Preventable Diseases, Department of Science and Innovation/National Research Foundation, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gaurav Kwatra
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH, USA.
- Department of Clinical Microbiology, Christian Medical College, Vellore, India.
| | - Alane Izu
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stephanie A Jones
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nkululeko J Mbele
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nwabisa Jafta
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Rushil Lala
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sharon Shalekoff
- A Division of the National Health Laboratory Service, Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Caroline T Tiemessen
- A Division of the National Health Laboratory Service, Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A Madhi
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Marta C Nunes
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Research Chair Initiative in Vaccine Preventable Diseases, Department of Science and Innovation/National Research Foundation, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Center of Excellence in Respiratory Pathogens, Hospices Civils de Lyon, and Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
9
|
Tapela K, Prah DA, Tetteh B, Nuokpem F, Dosoo D, Coker A, Kumi-Ansah F, Amoako E, Assah KO, Kilba C, Nyakoe N, Quansah D, Languon S, Anyigba CA, Ansah F, Agyeman S, Owusu IA, Schneider K, Ampofo WK, Mutungi JK, Amegatcher G, Aniweh Y, Awandare GA, Quashie PK, Bediako Y. Cellular immune response to SARS-CoV-2 and clinical presentation in individuals exposed to endemic malaria. Cell Rep 2024; 43:114533. [PMID: 39052480 PMCID: PMC11372439 DOI: 10.1016/j.celrep.2024.114533] [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: 08/31/2023] [Revised: 10/17/2023] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
Ghana and other parts of West Africa have experienced lower COVID-19 mortality rates than other regions. This phenomenon has been hypothesized to be associated with previous exposure to infections such as malaria. This study investigated the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the influence of previous malaria exposure. Blood samples were collected from individuals with asymptomatic or symptomatic COVID-19 (n = 217). A variety of assays were used to characterize the SARS-CoV-2-specific immune response, and malaria exposure was quantified using Plasmodium falciparum ELISA. The study found evidence of attenuated immune responses to COVID-19 among asymptomatic individuals, with elevated proportions of non-classical monocytes and greater memory B cell activation. Symptomatic patients displayed higher P. falciparum-specific T cell recall immune responses, whereas asymptomatic individuals demonstrated elevated P. falciparum antibody levels. Summarily, this study suggests that P. falciparum exposure-associated immune modulation may contribute to reduced severity of SARS-CoV-2 infection among people living in malaria-endemic regions.
Collapse
Affiliation(s)
- Kesego Tapela
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Diana Ahu Prah
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Becky Tetteh
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Franklin Nuokpem
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Daniel Dosoo
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Amin Coker
- Accident and Emergency Unit, The Greater Accra Regional Hospital, Accra, Ghana
| | | | - Emmanuella Amoako
- Department of Pediatrics, Cape Coast Teaching Hospital, Cape Coast, Ghana; Yemaachi Biotech Inc., 222 Swaniker St., Accra, Ghana
| | - Kissi Ohene Assah
- Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Charlyne Kilba
- Department of Internal Medicine, Surgery, Pediatrics, and Emergency Medicine, Greater Accra Regional Hospital, Accra, Ghana
| | - Nancy Nyakoe
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Darius Quansah
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana; Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Sylvester Languon
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Claudia Adzo Anyigba
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Felix Ansah
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Seth Agyeman
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana; Yemaachi Biotech Inc., 222 Swaniker St., Accra, Ghana
| | - Irene Amoakoh Owusu
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Kristan Schneider
- Department of Mathematics, Hochschule Mittweida, University of Applied Sciences, Mittweida, Germany
| | - William K Ampofo
- Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Joe Kimanthi Mutungi
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Gloria Amegatcher
- Department of Medical Laboratory Science, School of Biomedical and Allied Sciences, University of Ghana, Accra, Ghana
| | - Yaw Aniweh
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; The Francis Crick Institute, 1 Midland Rd., London NW1 1AT, UK
| | - Gordon A Awandare
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Peter K Quashie
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; The Francis Crick Institute, 1 Midland Rd., London NW1 1AT, UK.
| | - Yaw Bediako
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Yemaachi Biotech Inc., 222 Swaniker St., Accra, Ghana; The Francis Crick Institute, 1 Midland Rd., London NW1 1AT, UK.
| |
Collapse
|
10
|
Grebe E, Stone M, Spencer BR, Akinseye A, Wright DJ, Di Germanio C, Bruhn R, Zurita KG, Contestable P, Green V, Lanteri MC, Saa P, Biggerstaff BJ, Coughlin MM, Kleinman S, Custer B, Jones JM, Busch MP. Detection of Nucleocapsid Antibodies Associated with Primary SARS-CoV-2 Infection in Unvaccinated and Vaccinated Blood Donors. Emerg Infect Dis 2024; 30:1621-1630. [PMID: 38981189 PMCID: PMC11286071 DOI: 10.3201/eid3008.240659] [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: 07/11/2024] Open
Abstract
Nucleocapsid antibody assays can be used to estimate SARS-CoV-2 infection prevalence in regions implementing spike-based COVID-19 vaccines. However, poor sensitivity of nucleocapsid antibody assays in detecting infection after vaccination has been reported. We derived a lower cutoff for identifying previous infections in a large blood donor cohort (N = 142,599) by using the Ortho VITROS Anti-SARS-CoV-2 Total-N Antibody assay, improving sensitivity while maintaining specificity >98%. We validated sensitivity in samples donated after self-reported swab-confirmed infections diagnoses. Sensitivity for first infections in unvaccinated donors was 98.1% (95% CI 98.0-98.2) and for infection after vaccination was 95.6% (95% CI 95.6-95.7) based on the standard cutoff. Regression analysis showed sensitivity was reduced in the Delta compared with Omicron period, in older donors, in asymptomatic infections, <30 days after infection, and for infection after vaccination. The standard Ortho N antibody threshold demonstrated good sensitivity, which was modestly improved with the revised cutoff.
Collapse
Affiliation(s)
| | | | - Bryan R. Spencer
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Akintunde Akinseye
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - David J. Wright
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Clara Di Germanio
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Roberta Bruhn
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Karla G. Zurita
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Paul Contestable
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Valerie Green
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Marion C. Lanteri
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Paula Saa
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Brad J. Biggerstaff
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Melissa M. Coughlin
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Steve Kleinman
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Brian Custer
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Jefferson M. Jones
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| | - Michael P. Busch
- Vitalant Research Institute, San Francisco, California, USA (E. Grebe, M. Stone, C. Di Germanio, R. Bruhn, K.G. Zurita, B. Custer, M.P. Busch)
- University of California, San Francisco (M. Stone, R. Bruhn, M.C. Lanteri, B. Custer, M.P. Busch)
- American Red Cross, Rockville, Maryland, USA (B.R. Spencer, P. Saa)
- Westat, Rockville (A. Akinseye, D. Wright); QuidelOrtho, Rochester, New York, USA (P. Contestable)
- Creative Testing Solutions, Tempe, Arizona, USA (V. Green, M.C. Lanteri)
- Centers for Disease Control and Prevention, Fort Collins, Colorado, USA (B.J. Biggerstaff)
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA (M.M. Coughlin, J.M. Jones)
- University of British Columbia, Vancouver, British Columbia, Canada (S. Kleinman)
| |
Collapse
|
11
|
Konuma T, Hamatani-Asakura M, Nagai E, Adachi E, Kato S, Isobe M, Monna-Oiwa M, Takahashi S, Yotsuyanagi H, Nannya Y. Cellular and humoral immunogenicity against SARS-CoV-2 vaccination or infection is associated with the memory phenotype of T- and B-lymphocytes in adult allogeneic hematopoietic cell transplant recipients. Int J Hematol 2024; 120:229-240. [PMID: 38842630 PMCID: PMC11284193 DOI: 10.1007/s12185-024-03802-3] [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: 10/06/2023] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
We conducted a cross-sectional study to evaluate cellular and humoral immunogenicity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination or infection and examine how lymphocyte subpopulations in peripheral blood correlate with cellular and humoral immunogenicity in adult allogeneic hematopoietic cell transplantation (HCT) recipients. The median period from SARS-CoV-2 vaccination or infection to sample collection was 110.5 days (range, 6-345 days). The median SARS-CoV-2 spike-specific antibody level was 1761 binding antibody units (BAU)/ml (range, 0 to > 11,360 BAU/ml). Enzyme-linked immunosorbent spot (ELISpot) assay of T cells stimulated with SARS-CoV-2 spike antigens showed that interferon-gamma (IFN-γ)-, interleukin-2 (IL-2)-, and IFN-γ + IL-2-producing T cells were present in 68.9%, 62.0%, and 56.8% of patients, respectively. The antibody level was significantly correlated with frequency of IL-2-producing T cells (P = 0.001) and IFN-γ + IL-2-producing T cells (P = 0.006) but not IFN-γ-producing T cells (P = 0.970). Absolute counts of CD8+ and CD4+ central memory T cells were higher in both IL-2- and IFN-γ + IL-2-producing cellular responders compared with non-responders. These data suggest that cellular and humoral immunogenicity against SARS-CoV-2 vaccination or infection is associated with the memory phenotype of T cells and B cells in adult allogeneic HCT recipients.
Collapse
Affiliation(s)
- Takaaki Konuma
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, Japan.
| | - Megumi Hamatani-Asakura
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Etsuko Nagai
- Department of Laboratory Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Eisuke Adachi
- Department of Infectious Diseases and Applied Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiko Kato
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Masamichi Isobe
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Maki Monna-Oiwa
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Satoshi Takahashi
- Division of Clinical Precision Research Platform, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Yotsuyanagi
- Department of Infectious Diseases and Applied Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yasuhito Nannya
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, Japan
| |
Collapse
|
12
|
Hernandez-Galicia G, Gomez-Morales L, Lopez-Bailon LU, Valdovinos-Torres H, Contreras-Ochoa CO, Díaz Benítez CE, Martinez-Barnetche J, Alpuche-Aranda C, Ortiz-Navarrete V. Presence of SARS-CoV-2-specific T cells before vaccination in the Mexican population. J Leukoc Biol 2024; 116:95-102. [PMID: 38717738 DOI: 10.1093/jleuko/qiae111] [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: 08/01/2023] [Revised: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 06/30/2024] Open
Abstract
The immune response to SARS-CoV-2 has been extensively studied following the pandemic outbreak in 2020; however, the presence of specific T cells against SARS-CoV-2 before vaccination has not been evaluated in Mexico. In this study, we estimated the frequency of T CD4+ and T CD8+ cells that exhibit a specific response to S (spike) and N (nucleocapsid) proteins in a Mexican population. We collected 78 peripheral blood samples from unvaccinated subjects, and the presence of antibodies against spike (RBD) and N protein was determined. Peripheral blood mononuclear cells were isolated and stimulated with a pool of S or N protein peptides (Wuhan-Hu-1 strain). IL-1β, IL-4, IL-6, IL-10, IL-2, IL-8, TNF-α, IFN-γ, and GM-CSF levels were quantified in the supernatant of the activated cells, and the cells were stained to assess the activation and memory phenotypes. Differential activation frequency dependent on serological status was observed in CD4+ cells but not in CD8+ cells. The predominantly activated population was the central memory T CD4+ cells. Only 10% of the population exhibited the same phenotype with respect to the response to nucleocapsid peptides. The cytokine profile differed between the S and N responses. S peptides induced a more proinflammatory response compared with the N peptides. In conclusion, in a Mexican cohort before vaccination, there was a significant response to the S and N SARS-CoV-2 proteins resulting from previous infections with seasonal coronaviruses or previous undetected exposure to SARS-CoV-2.
Collapse
Affiliation(s)
- Gabriela Hernandez-Galicia
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
| | - Luis Gomez-Morales
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
- Department of Immunology, National School of Biological Sciences, National Polytechnic Institute, Manuel Carpio and Plan de Ayala St, 11340, Mexico City, Mexico
| | - Luis Uriel Lopez-Bailon
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
- Department of Immunology, National School of Biological Sciences, National Polytechnic Institute, Manuel Carpio and Plan de Ayala St, 11340, Mexico City, Mexico
| | - Humberto Valdovinos-Torres
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Carla O Contreras-Ochoa
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Cinthya Estefhany Díaz Benítez
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Jesus Martinez-Barnetche
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Celia Alpuche-Aranda
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Vianney Ortiz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
| |
Collapse
|
13
|
Wang X, Zhang J, Liu M, Guo Y, Guo P, Yang X, Shang B, Li M, Tian J, Zhang T, Wang X, Jin R, Zhou J, Gao GF, Liu J. Nonconserved epitopes dominate reverse preexisting T cell immunity in COVID-19 convalescents. Signal Transduct Target Ther 2024; 9:160. [PMID: 38866784 PMCID: PMC11169541 DOI: 10.1038/s41392-024-01876-3] [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: 10/05/2023] [Revised: 04/30/2024] [Accepted: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
The herd immunity against SARS-CoV-2 is continuously consolidated across the world during the ongoing pandemic. However, the potential function of the nonconserved epitopes in the reverse preexisting cross-reactivity induced by SARS-CoV-2 to other human coronaviruses is not well explored. In our research, we assessed T cell responses to both conserved and nonconserved peptides shared by SARS-CoV-2 and SARS-CoV, identifying cross-reactive CD8+ T cell epitopes using enzyme-linked immunospot and intracellular cytokine staining assays. Then, in vitro refolding and circular dichroism were performed to evaluate the thermal stability of the HLA/peptide complexes. Lastly, single-cell T cell receptor reservoir was analyzed based on tetramer staining. Here, we discovered that cross-reactive T cells targeting SARS-CoV were present in individuals who had recovered from COVID-19, and identified SARS-CoV-2 CD8+ T cell epitopes spanning the major structural antigens. T cell responses induced by the nonconserved peptides between SARS-CoV-2 and SARS-CoV were higher and played a dominant role in the cross-reactivity in COVID-19 convalescents. Cross-T cell reactivity was also observed within the identified series of CD8+ T cell epitopes. For representative immunodominant peptide pairs, although the HLA binding capacities for peptides from SARS-CoV-2 and SARS-CoV were similar, the TCR repertoires recognizing these peptides were distinct. Our results could provide beneficial information for the development of peptide-based universal vaccines against coronaviruses.
Collapse
Affiliation(s)
- Xin Wang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China
| | - Jie Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, P.R. China
| | - Maoshun Liu
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuanyuan Guo
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China
| | - Peipei Guo
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China
| | - Xiaonan Yang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China
| | - Bingli Shang
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Min Li
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China
| | - Jinmin Tian
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ting Zhang
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xi Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, P.R. China
| | - Ronghua Jin
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, P.R. China
| | - Jikun Zhou
- Shijiazhuang Fifth Hospital, Shijiazhuang, 050011, China.
| | - George F Gao
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China.
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jun Liu
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China.
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
14
|
Zhou D, Zhao S, He K, Liu Q, Zhang F, Pu Z, Xiao L, Zhang L, Chen S, Qian X, Wu X, Shen Y, Yu L, Zhang H, Jin J, Xu M, Wang X, Zhu D, Xie Z, Xu X. Longitudinal dynamic single-cell mass cytometry analysis of peripheral blood mononuclear cells in COVID-19 patients within 6 months after viral RNA clearance. BMC Infect Dis 2024; 24:567. [PMID: 38844850 PMCID: PMC11157885 DOI: 10.1186/s12879-024-09464-0] [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: 09/09/2023] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
This study investigates the longitudinal dynamic changes in immune cells in COVID-19 patients over an extended period after recovery, as well as the interplay between immune cells and antibodies. Leveraging single-cell mass spectrometry, we selected six COVID-19 patients and four healthy controls, dissecting the evolving landscape within six months post-viral RNA clearance, alongside the levels of anti-spike protein antibodies. The T cell immunophenotype ascertained via single-cell mass spectrometry underwent validation through flow cytometry in 37 samples. Our findings illuminate that CD8 + T cells, gamma-delta (gd) T cells, and NK cells witnessed an increase, in contrast to the reduction observed in monocytes, B cells, and double-negative T (DNT) cells over time. The proportion of monocytes remained significantly elevated in COVID-19 patients compared to controls even after six-month. Subpopulation-wise, an upsurge manifested within various T effector memory subsets, CD45RA + T effector memory, gdT, and NK cells, whereas declines marked the populations of DNT, naive and memory B cells, and classical as well as non-classical monocytes. Noteworthy associations surfaced between DNT, gdT, CD4 + T, NK cells, and the anti-S antibody titer. This study reveals the changes in peripheral blood mononuclear cells of COVID-19 patients within 6 months after viral RNA clearance and sheds light on the interactions between immune cells and antibodies. The findings from this research contribute to a better understanding of immune transformations during the recovery from COVID-19 and offer guidance for protective measures against reinfection in the context of viral variants.
Collapse
Affiliation(s)
- Diwenxin Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Shuai Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Keting He
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Qiuhong Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Fen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Zhangya Pu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Lanlan Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Lingjian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Shangci Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Xiaohan Qian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Xiaoxin Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Yangfan Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Ling Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Huafen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Jiandi Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Min Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Xiaoyan Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Zhongyang Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China.
| | - Xiaowei Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China.
| |
Collapse
|
15
|
Suryawanshi P, Patil‐Takbhate B, Athavale P, Mirza S, Tripathy A, Kanitkar S, Shivnitwar S, Barthwal MS, Dole S, Chavan H, Jali P, Pawale S, Kakad D, Kakrani AL, Bhawalkar J, Gandhi M, Chaturvedi S, Karandikar M, Tripathy S. T-cell responses in COVID-19 survivors 6-8 months after infection: A longitudinal cohort study in Pune. Immun Inflamm Dis 2024; 12:e1238. [PMID: 38860770 PMCID: PMC11165687 DOI: 10.1002/iid3.1238] [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: 10/27/2023] [Revised: 02/26/2024] [Accepted: 03/20/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immune response is crucial for disease management, although diminishing immunity raises the possibility of reinfection. METHODS We examined the immunological response to SARS-CoV-2 in a cohort of convalescent COVID-19 patients in matched samples collected at 1 and 6-8 months after infection. The peripheral blood mononuclear cells were isolated from enrolled study participants and flow cytometry analysis was done to assess the lymphocyte subsets of naive, effector, central memory, and effector memory CD4+ or CD8+ T cells in COVID-19 patients at 1 and 6-8 months after infection. Immunophenotypic characterization of immune cell subsets was performed on individuals who were followed longitudinally for 1 month (n = 44) and 6-8 months (n = 25) after recovery from COVID infection. RESULTS We observed that CD4 +T cells in hospitalized SARS-CoV-2 patients tended to decrease, whereas CD8+ T cells steadily recovered after 1 month, while there was a sustained increase in the population of effector T cells and effector memory T cells. Furthermore, COVID-19 patients showed persistently low B cells and a small increase in the NK cell population. CONCLUSION Our findings show that T cell responses were maintained at 6-8 months after infection. This opens new pathways for further research into the long-term effects in COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
- Poonam Suryawanshi
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Bhagyashri Patil‐Takbhate
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Prachi Athavale
- Department of Microbiology, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Shahzad Mirza
- Department of Microbiology, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | | | - Shubhangi Kanitkar
- Department of General Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Sachin Shivnitwar
- Department of General Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Madhusudan S. Barthwal
- Department of Respiratory Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, Pimpri, (deemed to be University)PuneIndia
| | - Sachin Dole
- Department of Respiratory Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, Pimpri, (deemed to be University)PuneIndia
| | - Hanumant Chavan
- Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Priyanka Jali
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Sujata Pawale
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Dhanashree Kakad
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Arjun Lal Kakrani
- Department of General Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Jitendra Bhawalkar
- Department of Community Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Madhura Gandhi
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | | | - Mahesh Karandikar
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Srikanth Tripathy
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| |
Collapse
|
16
|
Yang G, Cao J, Qin J, Mei X, Deng S, Xia Y, Zhao J, Wang J, Luan T, Chen D, Huang P, Chen C, Sun X, Luo Q, Su J, Zhang Y, Zhong N, Wang Z. Initial COVID-19 severity influenced by SARS-CoV-2-specific T cells imprints T-cell memory and inversely affects reinfection. Signal Transduct Target Ther 2024; 9:141. [PMID: 38811527 PMCID: PMC11136975 DOI: 10.1038/s41392-024-01867-4] [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: 02/10/2024] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 05/31/2024] Open
Abstract
The immunoprotective components control COVID-19 disease severity, as well as long-term adaptive immunity maintenance and subsequent reinfection risk discrepancies across initial COVID-19 severity, remain unclarified. Here, we longitudinally analyzed SARS-CoV-2-specific immune effectors during the acute infection and convalescent phases of 165 patients with COVID-19 categorized by severity. We found that early and robust SARS-CoV-2-specific CD4+ and CD8+ T cell responses ameliorate disease progression and shortened hospital stay, while delayed and attenuated virus-specific CD8+ T cell responses are prominent severe COVID-19 features. Delayed antiviral antibody generation rather than titer level associates with severe outcomes. Conversely, initial COVID-19 severity imprints the long-term maintenance of SARS-CoV-2-specific adaptive immunity, demonstrating that severe convalescents exhibited more sustained virus-specific antibodies and memory T cell responses compared to mild/moderate counterparts. Moreover, initial COVID-19 severity inversely correlates with SARS-CoV-2 reinfection risk. Overall, our study unravels the complicated interaction between temporal characteristics of virus-specific T cell responses and COVID-19 severity to guide future SARS-CoV-2 wave management.
Collapse
Affiliation(s)
- Gang Yang
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| | - Jinpeng Cao
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jian Qin
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
| | - Xinyue Mei
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Shidong Deng
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yingjiao Xia
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
| | - Jun Zhao
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
| | - Junxiang Wang
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Tao Luan
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| | - Daxiang Chen
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Peiyu Huang
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Cheng Chen
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
| | - Xi Sun
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Qi Luo
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jie Su
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yunhui Zhang
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China.
| | - Nanshan Zhong
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China.
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China.
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| | - Zhongfang Wang
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China.
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
17
|
Weisbrod L, Capriotti L, Hofmann M, Spieler V, Dersch H, Voedisch B, Schmidt P, Knake S. FASTMAP-a flexible and scalable immunopeptidomics pipeline for HLA- and antigen-specific T-cell epitope mapping based on artificial antigen-presenting cells. Front Immunol 2024; 15:1386160. [PMID: 38779658 PMCID: PMC11109385 DOI: 10.3389/fimmu.2024.1386160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
The study of peptide repertoires presented by major histocompatibility complex (MHC) molecules and the identification of potential T-cell epitopes contribute to a multitude of immunopeptidome-based treatment approaches. Epitope mapping is essential for the development of promising epitope-based approaches in vaccination as well as for innovative therapeutics for autoimmune diseases, infectious diseases, and cancer. It also plays a critical role in the immunogenicity assessment of protein therapeutics with regard to safety and efficacy concerns. The main challenge emerges from the highly polymorphic nature of the human leukocyte antigen (HLA) molecules leading to the requirement of a peptide mapping strategy for a single HLA allele. As many autoimmune diseases are linked to at least one specific antigen, we established FASTMAP, an innovative strategy to transiently co-transfect a single HLA allele combined with a disease-specific antigen into a human cell line. This approach allows the specific identification of HLA-bound peptides using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Using FASTMAP, we found a comparable spectrum of endogenous peptides presented by the most frequently expressed HLA alleles in the world's population compared to what has been described in literature. To ensure a reliable peptide mapping workflow, we combined the HLA alleles with well-known human model antigens like coagulation factor VIII, acetylcholine receptor subunit alpha, protein structures of the SARS-CoV-2 virus, and myelin basic protein. Using these model antigens, we have been able to identify a broad range of peptides that are in line with already published and in silico predicted T-cell epitopes of the specific HLA/model antigen combination. The transient co-expression of a single affinity-tagged MHC molecule combined with a disease-specific antigen in a human cell line in our FASTMAP pipeline provides the opportunity to identify potential T-cell epitopes/endogenously processed MHC-bound peptides in a very cost-effective, fast, and customizable system with high-throughput potential.
Collapse
Affiliation(s)
- Luisa Weisbrod
- Recombinant Protein Discovery, CSL Innovation GmbH, Marburg, Germany
| | - Luigi Capriotti
- Analytical Biochemistry, Research and Development, CSL Behring AG, Bern, Switzerland
| | - Marco Hofmann
- Recombinant Protein Discovery, CSL Innovation GmbH, Marburg, Germany
| | - Valerie Spieler
- Recombinant Protein Discovery, CSL Innovation GmbH, Marburg, Germany
| | - Herbert Dersch
- Recombinant Protein Discovery, CSL Innovation GmbH, Marburg, Germany
| | - Bernd Voedisch
- Recombinant Protein Discovery, CSL Innovation GmbH, Marburg, Germany
| | - Peter Schmidt
- Protein Biochemistry, Bio21 Institute, CSL Limited, Parkville, VIC, Australia
| | - Susanne Knake
- Department of Neurology, Epilepsy Center Hessen, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
18
|
Baimakanova GE, Samsonova M, Chernyaev AL, Kontorschikov AS, Belevskiy AS. [Clinical and morphological features of lung injury long-term after SARS-CoV-2 recovery]. TERAPEVT ARKH 2024; 96:218-227. [PMID: 38713035 DOI: 10.26442/00403660.2024.03.202647] [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: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 05/08/2024]
Abstract
AIM To study the clinical and histological profile of lung tissue in patients with persistent pulmonary disease, respiratory symptoms and CT findings after SARS-CoV-2 infection. MATERIALS AND METHODS The study included 15 patients (7 females and 8 males) with a mean age of 57.7 years. All patients underwent laboratory tests, chest computed tomography, echocardiography, and pulmonary function tests. Pulmonary tissue and bronchoalveolar lavage samples were obtained by fibrobronchoscopy, transbronchial forceps (2 patients), and lung cryobiopsy (11 patients); open biopsy was performed in 2 patients. Cellular composition, herpesvirus DNA, SARS-CoV-2, Mycobacterium tuberculosis complex, galactomannan optical density index, and bacterial and fungal microflora growth were determined in bronchoalveolar lavage. SARS-CoV-2 was also identified in samples from the nasal mucosa, throat and feces using a polymerase chain reaction. RESULTS The results showed no true pulmonary fibrosis in patients recovered from SARS-CoV-2 infection with persistent respiratory symptoms, functional impairment, and CT findings after SARS-CoV-2 infection. The observed changes comply with the current and/or resolving infection and inflammatory process. CONCLUSION Thus, no true pulmonary fibrosis was found in patients after SARS-CoV-2 infection with persistent respiratory symptoms, functional impairment, and CT findings. The observed changes comply with the current and/or resolving infection and inflammatory process.
Collapse
Affiliation(s)
| | - M Samsonova
- Loginov Moscow Clinical Scientific Center
- Research Institute of Pulmonology
| | - A L Chernyaev
- Research Institute of Pulmonology
- Petrovsky National Research Centre of Surgery
- Pirogov Russian National Research Medical University
| | | | - A S Belevskiy
- Pirogov Russian National Research Medical University
| |
Collapse
|
19
|
Montmaneix-Engels F, Dimeglio C, Staes L, Da Silva I, Porcheron M, Jougla I, Hérin F, Izopet J. Study of the cellular and humoral immune responses to SARS-CoV-2 vaccination. Heliyon 2024; 10:e29116. [PMID: 38601689 PMCID: PMC11004869 DOI: 10.1016/j.heliyon.2024.e29116] [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: 01/11/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/12/2024] Open
Abstract
Our understanding of cellular immunity in response to COVID-19 infection or vaccination is limited because of less commonly used techniques. We investigated both the cellular and humoral immune responses before and after the administration of a third dose of the SARS-CoV-2 vaccine among a group of healthcare workers. Cellular immunity was evaluated using the VIDAS interferon-gamma (IFNγ) RUO test, which enables automated measurement of IFNγ levels after stimulating peripheral blood lymphocytes. Booster doses significantly enhanced both cellular and humoral immunity. Concerning cellular response, the booster dose increased the percentage of positive IFNγ release assay (IGRA) results but no difference in IFNγ release was found. The cellular response was not associated with protection against SARS-CoV-2 infection. Interestingly, vaccinated and infected healthcare workers exhibited the highest levels of anti-spike and neutralizing antibodies. In conclusion, the IGRA is a simple method for measuring cellular immune responses after vaccination. However, its usefulness as a complement to the study of humoral responses is yet to be demonstrated in future research.
Collapse
Affiliation(s)
- Faustine Montmaneix-Engels
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- Toulouse III Paul Sabatier University, 31062, Toulouse, France
| | - Chloé Dimeglio
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Laeticia Staes
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Isabelle Da Silva
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Marion Porcheron
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Isabelle Jougla
- Occupational Diseases Department, Toulouse University Hospital, 31000, Toulouse, France
| | - Fabrice Hérin
- Occupational Diseases Department, Toulouse University Hospital, 31000, Toulouse, France
- UMR1295, Joint Research Unit INSERM- University Toulouse III Paul Sabatier, Centre for Epidemiology and Research in Population Health Unit (CERPOP), 31000, Toulouse, France
| | - Jacques Izopet
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- Toulouse III Paul Sabatier University, 31062, Toulouse, France
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| |
Collapse
|
20
|
Wu N, Zhang J, Shen Y, Zhang X, Zhou J, Wu Y, Li E, Meng X, Chuai X, Chiu S, Wang Y. A potential bivalent mRNA vaccine candidate protects against both RSV and SARS-CoV-2 infections. Mol Ther 2024; 32:1033-1047. [PMID: 38341613 PMCID: PMC11163217 DOI: 10.1016/j.ymthe.2024.02.011] [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: 08/31/2023] [Revised: 12/29/2023] [Accepted: 02/07/2024] [Indexed: 02/12/2024] Open
Abstract
As the world continues to confront severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), respiratory syncytial virus (RSV) is also causing severe respiratory illness in millions of infants, elderly individuals, and immunocompromised people globally. Exacerbating the situation is the fact that co-infection with multiple viruses is occurring, something which has greatly increased the clinical severity of the infections. Thus, our team developed a bivalent vaccine that delivered mRNAs encoding SARS-CoV-2 Omicron spike (S) and RSV fusion (F) proteins simultaneously, SF-LNP, which induced S and F protein-specific binding antibodies and cellular immune responses in BALB/c mice. Moreover, SF-LNP immunization effectively protected BALB/c mice from RSV infection and hamsters from SARS-CoV-2 Omicron infection. Notably, our study pointed out the antigenic competition problem of bivalent vaccines and provided a solution. Overall, our results demonstrated the potential of preventing two infectious diseases with a single vaccine and provided a paradigm for the subsequent design of multivalent vaccines.
Collapse
Affiliation(s)
- Namei Wu
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, P.R. China; School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China
| | - Jiachen Zhang
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China
| | - Yanqiong Shen
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China
| | - Xinghai Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430062, P.R. China
| | - Jinge Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430062, P.R. China
| | - Yan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430062, P.R. China
| | - Entao Li
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China
| | - Xiaoming Meng
- School of Pharmacy, Anhui Medical University, Hefei 230027, P.R. China
| | - Xia Chuai
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430062, P.R. China.
| | - Sandra Chiu
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China.
| | - Yucai Wang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, P.R. China; School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China; RNAlfa Biotech, Hefei 230088, P.R. China.
| |
Collapse
|
21
|
Kim SH, Kim Y, Jeon S, Park U, Kang JI, Jeon K, Kim HR, Oh S, Rhee JY, Choi JP, Park WB, Park SW, Yang JS, Lee JY, Kang J, Shin HS, Kim Y, Kim S, Kim YS, Lim DG, Cho NH. Rise in broadly cross-reactive adaptive immunity against human β-coronaviruses in MERS-recovered patients during the COVID-19 pandemic. SCIENCE ADVANCES 2024; 10:eadk6425. [PMID: 38416834 PMCID: PMC10901372 DOI: 10.1126/sciadv.adk6425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 01/23/2024] [Indexed: 03/01/2024]
Abstract
To develop a universal coronavirus (CoV) vaccine, long-term immunity against multiple CoVs, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, Middle East respiratory syndrome (MERS)-CoV, and future CoV strains, is crucial. Following the 2015 Korean MERS outbreak, we conducted a long-term follow-up study and found that although neutralizing antibodies and memory T cells against MERS-CoV declined over 5 years, some recovered patients exhibited increased antibody levels during the COVID-19 pandemic. This likely resulted from cross-reactive immunity induced by SARS-CoV-2 vaccines or infections. A significant correlation in antibody responses across various CoVs indicates shared immunogenic epitopes. Two epitopes-the spike protein's stem helix and intracellular domain-were highly immunogenic after MERS-CoV infection and after SARS-CoV-2 vaccination or infection. In addition, memory T cell responses, especially polyfunctional CD4+ T cells, were enhanced during the pandemic, correlating significantly with MERS-CoV spike-specific antibodies and neutralizing activity. Therefore, incorporating these cross-reactive and immunogenic epitopes into pan-CoV vaccine formulations may facilitate effective vaccine development.
Collapse
Affiliation(s)
- So-Hee Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Yuri Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Institute of Endemic Disease, Seoul National University Medical Research, Seoul 03080, Republic of Korea
| | - Sangeun Jeon
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea
| | - Uni Park
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Ju-Il Kang
- Institute of Endemic Disease, Seoul National University Medical Research, Seoul 03080, Republic of Korea
| | - Kyeongseok Jeon
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Hye-Ran Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Songhyeok Oh
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Ji-Young Rhee
- Division of Infectious Diseases, Department of Medicine, Dankook University College of Medicine, Cheonan 31116, Republic of Korea
| | - Jae-Phil Choi
- Department of Internal Medicine, Seoul Medical Center, Seoul 02053, Republic of Korea
| | - Wan Beom Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sang Won Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jeong-Sun Yang
- Center for Emerging Virus Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju 28159, Republic of Korea
| | - Joo-Yeon Lee
- Center for Emerging Virus Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju 28159, Republic of Korea
| | - Jihye Kang
- Translational Research Center, Research Institute of Public Health, National Medical Center, Seoul 04564, Republic of Korea
| | - Hyoung-Shik Shin
- Division of Infectious Diseases, Department of Internal Medicine, Daejeon Eulji Medical Center, Eulji University School of Medicine, Daejeon 34824, Republic of Korea
| | - Yeonjae Kim
- Center for Infectious Diseases, National Medical Center, Seoul 04564, Republic of Korea
| | - Seungtaek Kim
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea
| | - Yeon-Sook Kim
- Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Dong-Gyun Lim
- Translational Research Center, Research Institute of Public Health, National Medical Center, Seoul 04564, Republic of Korea
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Institute of Endemic Disease, Seoul National University Medical Research, Seoul 03080, Republic of Korea
- Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do 13620, Republic of Korea
| |
Collapse
|
22
|
Bohmwald K, Diethelm-Varela B, Rodríguez-Guilarte L, Rivera T, Riedel CA, González PA, Kalergis AM. Pathophysiological, immunological, and inflammatory features of long COVID. Front Immunol 2024; 15:1341600. [PMID: 38482000 PMCID: PMC10932978 DOI: 10.3389/fimmu.2024.1341600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/09/2024] [Indexed: 04/12/2024] Open
Abstract
The COVID-19 pandemic continues to cause severe global disruption, resulting in significant excess mortality, overwhelming healthcare systems, and imposing substantial social and economic burdens on nations. While most of the attention and therapeutic efforts have concentrated on the acute phase of the disease, a notable proportion of survivors experience persistent symptoms post-infection clearance. This diverse set of symptoms, loosely categorized as long COVID, presents a potential additional public health crisis. It is estimated that 1 in 5 COVID-19 survivors exhibit clinical manifestations consistent with long COVID. Despite this prevalence, the mechanisms and pathophysiology of long COVID remain poorly understood. Alarmingly, evidence suggests that a significant proportion of cases within this clinical condition develop debilitating or disabling symptoms. Hence, urgent priority should be given to further studies on this condition to equip global public health systems for its management. This review provides an overview of available information on this emerging clinical condition, focusing on the affected individuals' epidemiology, pathophysiological mechanisms, and immunological and inflammatory profiles.
Collapse
Affiliation(s)
- Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Benjamín Diethelm-Varela
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Linmar Rodríguez-Guilarte
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Thomas Rivera
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
23
|
Suntronwong N, Kanokudom S, Auphimai C, Thongmee T, Assawakosri S, Vichaiwattana P, Yorsaeng R, Duangchinda T, Chantima W, Pakchotanon P, Nilyanimit P, Srimuan D, Thatsanathorn T, Sudhinaraset N, Wanlapakorn N, Poovorawan Y. Long-Term Dynamic Changes in Hybrid Immunity over Six Months after Inactivated and Adenoviral Vector Vaccination in Individuals with Previous SARS-CoV-2 Infection. Vaccines (Basel) 2024; 12:180. [PMID: 38400163 PMCID: PMC10891631 DOI: 10.3390/vaccines12020180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 01/30/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Numerous studies have largely focused on short-term immunogenicity in recovered individuals post mRNA vaccination. However, understanding the long-term durability, particularly in inactivated and adenoviral vectored vaccines, remains limited. We evaluated antibody responses, omicron variant neutralization, and IFN-γ responses in 119 previously infected individuals vaccinated with CoronaVac or ChAdOx1 up to six months post-vaccination. Both vaccines elicited robust immune responses in recovered individuals, surpassing those who were infection-naïve, and these persisted above pre-vaccination levels for six months. However, antibody levels declined over time (geometric mean ratio (GMR) = 0.52 for both vaccines). Notably, neutralizing activities against omicron declined faster in ChAdOx1 (GMR = 0.6) compared to CoronaVac recipients (GMR = 1.03). While the first dose of ChAdOx1 adequately induced immune responses in recovered individuals, a second dose demonstrated advantages in omicron variant neutralization and slower decline. Although both vaccines induced T cell responses, the median IFN-γ level at six months returned to pre-vaccination levels. However, more individuals exhibited reactive T cell responses. Extending the interval (13-15 months) between infection and vaccination could enhance antibody levels and broaden neutralization. Together, these findings demonstrate a robust humoral and cellular response that was sustained for at least six months after vaccination, thus guiding optimal vaccination strategies based on prior infection and vaccine platforms.
Collapse
Affiliation(s)
- Nungruthai Suntronwong
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
| | - Sitthichai Kanokudom
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
- Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Chompoonut Auphimai
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
| | - Thanunrat Thongmee
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
| | - Suvichada Assawakosri
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
- Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Preeyaporn Vichaiwattana
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
| | - Ritthideach Yorsaeng
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
| | - Thaneeya Duangchinda
- Molecular Biology of Dengue and Flaviviruses Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Development Agency, NSTDA, Pathum Thani 12120, Thailand; (T.D.); (P.P.)
| | - Warangkana Chantima
- Division of Dengue Hemorrhagic Fever Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Pattarakul Pakchotanon
- Molecular Biology of Dengue and Flaviviruses Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Development Agency, NSTDA, Pathum Thani 12120, Thailand; (T.D.); (P.P.)
| | - Pornjarim Nilyanimit
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
| | - Donchida Srimuan
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
| | - Thaksaporn Thatsanathorn
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
| | - Natthinee Sudhinaraset
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
| | - Nasamon Wanlapakorn
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (S.K.); (C.A.); (T.T.); (S.A.); (P.V.); (R.Y.); (P.N.); (D.S.); (T.T.); (N.S.); (N.W.)
- The Royal Society of Thailand (FRS(T)), Sanam Sueapa, Dusit, Bangkok 10330, Thailand
| |
Collapse
|
24
|
Benede N, Tincho MB, Walters A, Subbiah V, Ngomti A, Baguma R, Butters C, Hahnle L, Mennen M, Skelem S, Adriaanse M, Facey-Thomas H, Scott C, Day J, Spracklen TF, van Graan S, Balla SR, Moyo-Gwete T, Moore PL, MacGinty R, Botha M, Workman L, Johnson M, Goldblatt D, Zar HJ, Ntusi NA, Zühlke L, Webb K, Riou C, Burgers WA, Keeton RS. Distinct T cell polyfunctional profile in SARS-CoV-2 seronegative children associated with endemic human coronavirus cross-reactivity. iScience 2024; 27:108728. [PMID: 38235336 PMCID: PMC10792240 DOI: 10.1016/j.isci.2023.108728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/19/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024] Open
Abstract
SARS-CoV-2 infection in children typically results in asymptomatic or mild disease. There is a paucity of studies on SARS-CoV-2 antiviral immunity in African children. We investigated SARS-CoV-2-specific T cell responses in 71 unvaccinated asymptomatic South African children who were seropositive or seronegative for SARS-CoV-2. SARS-CoV-2-specific CD4+ T cell responses were detectable in 83% of seropositive and 60% of seronegative children. Although the magnitude of the CD4+ T cell response did not differ significantly between the two groups, their functional profiles were distinct, with SARS-CoV-2 seropositive children exhibiting a higher proportion of polyfunctional T cells compared to their seronegative counterparts. The frequency of SARS-CoV-2-specific CD4+ T cells in seronegative children was associated with the endemic human coronavirus (HCoV) HKU1 IgG response. Overall, the presence of SARS-CoV-2-responding T cells in seronegative children may result from cross-reactivity to endemic coronaviruses and could contribute to the relative protection from disease observed in SARS-CoV-2-infected children.
Collapse
Affiliation(s)
- Ntombi Benede
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Marius B. Tincho
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Avril Walters
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Vennesa Subbiah
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Amkele Ngomti
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Richard Baguma
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Claire Butters
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
| | - Lina Hahnle
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Mathilda Mennen
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Sango Skelem
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Marguerite Adriaanse
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Heidi Facey-Thomas
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Christiaan Scott
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
| | - Jonathan Day
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
| | - Timothy F. Spracklen
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
- South African Medical Research Council, Francie Van Zijl Drive, Parow Cape Town, South Africa
| | - Strauss van Graan
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Sashkia R. Balla
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Thandeka Moyo-Gwete
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Penny L. Moore
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Rae MacGinty
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Maresa Botha
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Lesley Workman
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Marina Johnson
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | - David Goldblatt
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | - Heather J. Zar
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Ntobeko A.B. Ntusi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Liesl Zühlke
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
- South African Medical Research Council, Francie Van Zijl Drive, Parow Cape Town, South Africa
| | - Kate Webb
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
- Crick African Network, The Francis Crick Institute, London, UK
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Wendy A. Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Roanne S. Keeton
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| |
Collapse
|
25
|
Binayke A, Zaheer A, Vishwakarma S, Singh S, Sharma P, Chandwaskar R, Gosain M, Raghavan S, Murugesan DR, Kshetrapal P, Thiruvengadam R, Bhatnagar S, Pandey AK, Garg PK, Awasthi A. A quest for universal anti-SARS-CoV-2 T cell assay: systematic review, meta-analysis, and experimental validation. NPJ Vaccines 2024; 9:3. [PMID: 38167915 PMCID: PMC10762233 DOI: 10.1038/s41541-023-00794-9] [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: 07/02/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Measuring SARS-CoV-2-specific T cell responses is crucial to understanding an individual's immunity to COVID-19. However, high inter- and intra-assay variability make it difficult to define T cells as a correlate of protection against COVID-19. To address this, we performed systematic review and meta-analysis of 495 datasets from 94 original articles evaluating SARS-CoV-2-specific T cell responses using three assays - Activation Induced Marker (AIM), Intracellular Cytokine Staining (ICS), and Enzyme-Linked Immunospot (ELISPOT), and defined each assay's quantitative range. We validated these ranges using samples from 193 SARS-CoV-2-exposed individuals. Although IFNγ ELISPOT was the preferred assay, our experimental validation suggested that it under-represented the SARS-CoV-2-specific T cell repertoire. Our data indicate that a combination of AIM and ICS or FluoroSpot assay would better represent the frequency, polyfunctionality, and compartmentalization of the antigen-specific T cell responses. Taken together, our results contribute to defining the ranges of antigen-specific T cell assays and propose a choice of assay that can be employed to better understand the cellular immune response against viral diseases.
Collapse
Affiliation(s)
- Akshay Binayke
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India
- Jawaharlal Nehru University, New Delhi, India
| | - Aymaan Zaheer
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Siddhesh Vishwakarma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Savita Singh
- Translational Health Science and Technology Institute, Faridabad, India
| | - Priyanka Sharma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Rucha Chandwaskar
- Department of Microbiology, AMITY University Rajasthan, Jaipur, India
| | - Mudita Gosain
- Translational Health Science and Technology Institute, Faridabad, India
| | | | | | | | - Ramachandran Thiruvengadam
- Translational Health Science and Technology Institute, Faridabad, India
- Pondicherry Institute of Medical Sciences, Puducherry, India
| | | | | | - Pramod Kumar Garg
- Translational Health Science and Technology Institute, Faridabad, India
- All India Institute of Medical Sciences, New Delhi, India
| | - Amit Awasthi
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India.
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India.
| |
Collapse
|
26
|
Calderilla-Barbosa L, Flores-Sevilla JL. CYTOSPOT: Intracellular Staining and Detection of Cytokines by Flow Cytometer, an Alternative to LYMESPOT. Methods Mol Biol 2024; 2742:91-98. [PMID: 38165617 DOI: 10.1007/978-1-0716-3561-2_7] [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: 01/04/2024]
Abstract
Intracellular cytokine staining is a versatile technique used to analyze cytokine production in individual cells by flow cytometry. This methodology has the specific advantage of enabling the simultaneous assessment of multiple phenotypic, differentiation, and functional parameters pertaining to responding T cells. This methodology applied after short-term culture of cells, followed by fixation and permeabilization make this technique ideal for the assessment of T-cell immune responses induced by different challenges. Here we describe an intracellular staining method followed by flow cytometry after cell stimulation with immune-relevant antigens for Lyme disease.
Collapse
|
27
|
Diego JGB, Singh G, Jangra S, Handrejk K, Laporte M, Chang LA, El Zahed SS, Pache L, Chang MW, Warang P, Aslam S, Mena I, Webb BT, Benner C, García-Sastre A, Schotsaert M. Breakthrough infections by SARS-CoV-2 variants boost cross-reactive hybrid immune responses in mRNA-vaccinated Golden Syrian hamsters. PLoS Pathog 2024; 20:e1011805. [PMID: 38198521 PMCID: PMC10805310 DOI: 10.1371/journal.ppat.1011805] [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: 06/03/2023] [Revised: 01/23/2024] [Accepted: 11/06/2023] [Indexed: 01/12/2024] Open
Abstract
Hybrid immunity (vaccination + natural infection) to SARS-CoV-2 provides superior protection to re-infection. We performed immune profiling studies during breakthrough infections in mRNA-vaccinated hamsters to evaluate hybrid immunity induction. The mRNA vaccine, BNT162b2, was dosed to induce binding antibody titers against ancestral spike, but inefficient serum virus neutralization of ancestral SARS-CoV-2 or variants of concern (VoCs). Vaccination reduced morbidity and controlled lung virus titers for ancestral virus and Alpha but allowed breakthrough infections in Beta, Delta and Mu-challenged hamsters. Vaccination primed for T cell responses that were boosted by infection. Infection back-boosted neutralizing antibody responses against ancestral virus and VoCs. Hybrid immunity resulted in more cross-reactive sera, reflected by smaller antigenic cartography distances. Transcriptomics post-infection reflects both vaccination status and disease course and suggests a role for interstitial macrophages in vaccine-mediated protection. Therefore, protection by vaccination, even in the absence of high titers of neutralizing antibodies in the serum, correlates with recall of broadly reactive B- and T-cell responses.
Collapse
Affiliation(s)
- Juan García-Bernalt Diego
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca-Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sonia Jangra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kim Handrejk
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Manon Laporte
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lauren A. Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sara S. El Zahed
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lars Pache
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Max W. Chang
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sadaf Aslam
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Brett T. Webb
- Department of Veterinary Sciences, University of Wyoming, Laramie, Wyoming, United States of America
| | - Christopher Benner
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
28
|
Petersen MS, Pérez-Alós L, í Kongsstovu SK, Eliasen EH, Hansen CB, Larsen S, Hansen JL, Bayarri-Olmos R, Fjallsbak JP, Weihe P, Garred P. Diverging humoral and cellular immune responses due to Omicron-a national study from the Faroe Islands. Microbiol Spectr 2023; 11:e0086523. [PMID: 37909772 PMCID: PMC10714973 DOI: 10.1128/spectrum.00865-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/30/2023] [Indexed: 11/03/2023] Open
Abstract
IMPORTANCE The immunity following infection and vaccination with the SARS-CoV-2 Omicron variant is poorly understood. We investigated immunity assessed with antibody and T-cell responses under different scenarios in vaccinated and unvaccinated individuals with and without Omicron infection. We found that the humoral response was higher among vaccinated-naïve than unvaccinated convalescent. Unvaccinated with and without infection had comparable low humoral responses, whereas vaccinated with a second or third dose, independent of infection status, had increasingly higher levels. Only a minor fraction of unvaccinated individuals had detectable humoral responses following Omicron infection, while almost all had positive T-cell responses. In conclusion, primary Omicron infection mounts a low humoral immune response, enhanced by prior vaccination. Omicron infection induced a robust T-cell response in both unvaccinated and vaccinated, demonstrating that immune evasion of primary Omicron infection affects humoral immunity more than T-cell immunity.
Collapse
Affiliation(s)
- Maria Skaalum Petersen
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Eina Hansen Eliasen
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
| | - Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Sólrun Larsen
- Chief Medical Officer Office, Tórshavn, Faroe Islands
| | | | - Rafael Bayarri-Olmos
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Pál Weihe
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
29
|
Wu CY, Tseng YC, Kao SE, Wu LY, Hou JT, Yang YC, Hsiao PW, Chen JR. Monoglycosylated SARS-CoV-2 receptor binding domain fused with HA stem-scaffolded protein vaccine confers broad protective immunity against SARS-CoV-2 and influenza viruses. Antiviral Res 2023; 220:105759. [PMID: 37984568 DOI: 10.1016/j.antiviral.2023.105759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
The SARS-CoV-2 and influenza pandemics have posed a devastating threat to global public health. The best strategy for preventing the further spread of these respiratory viruses worldwide is to administer a vaccine capable of targeting both viruses. Here, we show that a novel monoglycosylated vaccine designed based on the influenza virus HAstem conserved domain fused with the SARS-CoV-2 spike-RBD domain (HSSRmg) can present proper antigenicity that elicits sufficient neutralization efficacy against various SARS-CoV-2 variants while simultaneously providing broad protection against H1N1 viruses in mice. Compared with the fully glycosylated HSSR (HSSRfg), HSSRmg induced higher ELISA titers targeting HAstem and spike-RBD and exhibited significantly enhanced neutralization activity against the Wuhan pseudovirus. The enhanced immune responses raised by JR300-adjuvanted HSSRmg compared to HSSRmg alone include more anti-HAstem and anti-spike-RBD antibodies that provide cross-protection against H1N1 challenges and cross-neutralization of SARS-CoV-2 pseudoviruses. Furthermore, the enhanced immune response raised by JR300-adjuvanted-HSSRmg skews toward a more balanced Th1/Th2 response than that raised by HSSRmg alone. Notably, HSSRmg elicited more plasma B cells and memory B cells, and higher IL-4 and IFN-γ cytokine immune responses than spike (S-2P) in mice with preexisting influenza-specific immunity, suggesting that B-cell activation most likely occurs through CD4+ T-cell stimulation. This study demonstrated that HSSRmg produced using a monoglycosylation process and combined with the JR300 adjuvant elicits superior cross-strain immune responses against SARS-CoV-2 and influenza viruses in mice compared with S-2P. JR300-adjuvanted HSSRmg has great potential as a coronavirus-influenza vaccine that provides dual protection against SARS-CoV-2 and influenza infections.
Collapse
Affiliation(s)
| | | | - Shao-En Kao
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan
| | - Li-Yang Wu
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan
| | - Jen-Tzu Hou
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan
| | - Yu-Chih Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | | |
Collapse
|
30
|
Bansal S, Fleming T, Canez J, Maine GN, Bharat A, Walia R, Tokman S, Smith MA, Tiffany B, Bremner RM, Mohanakumar T. Immune responses of lung transplant recipients against SARS-CoV-2 and common respiratory coronaviruses: Evidence for pre-existing cross-reactive immunity. Transpl Immunol 2023; 81:101940. [PMID: 37866672 PMCID: PMC11019873 DOI: 10.1016/j.trim.2023.101940] [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: 08/04/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Humoral and cellular immune responses to SARS-CoV-2 and other coronaviruses in lung transplant recipients are unknown. We measured antibodies and T cell responses against the SARS-CoV-2 spike S2 and nucleocapsid antigens and spike antigens from common respiratory coronaviruses (229E, NL63, OC43, and HKU1) after vaccination or infection of LTxRs. 148 LTxRs from single center were included in this study: 98 after vaccination and 50 following SARS-CoV-2 infection. Antibodies were quantified by enzyme-linked immunosorbent assay. The frequency of T cells secreting IL2, IL4, IL10, IL17, TNFα, and IFNγ were enumerated by enzyme-linked immunospot assay. Our results have shown the development of antibodies to SARS-CoV-2 spike protein in infected LTxRs (39/50) and vaccinated LTxRs (52/98). Vaccinated LTxRs had higher number of T cells producing TNFα but less cells producing IFNγ than infected LTxRs in response to the nucleocapsid antigen and other coronavirus spike antigens. We didn't find correlation between the development of antibodies and cellular immune responses against the SARS-CoV-2 spike protein after vaccination. Instead, LTxRs have pre-existing cellular immunity to common respiratory coronaviruses, leading to cross-reactive immunity against SARS-CoV-2 which likely will provide protection against SARS-Cov-2 infection.
Collapse
Affiliation(s)
- Sandhya Bansal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Timothy Fleming
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Jesse Canez
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Gabriel N Maine
- Department of Pathology and Laboratory Medicine, Royal Oak, Beaumont Health, MI, USA
| | | | - Rajat Walia
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Sofya Tokman
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Michael A Smith
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Brian Tiffany
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Ross M Bremner
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - T Mohanakumar
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA.
| |
Collapse
|
31
|
Chen C, Wang X, Zhang Z. Humoral and cellular immunity against diverse SARS-CoV-2 variants. J Genet Genomics 2023; 50:934-947. [PMID: 37865193 DOI: 10.1016/j.jgg.2023.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 10/23/2023]
Abstract
Since the outbreak of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in late 2019, the virus has rapidly spread worldwide. This has led to an unprecedented global pandemic, marked by millions of COVID-19 cases and a significant number of fatalities. Over a relatively short period, several different vaccine platforms are developed and deployed for use globally to curb the pandemic. However, the genome of SARS-CoV-2 continuously undergoes mutation and/or recombination, resulting in the emergence of several variants of concern (VOC). These VOCs can elevate viral transmission and evade the neutralizing antibodies induced by vaccines, leading to reinfections. Understanding the impact of the SARS-CoV-2 genomic mutation on viral pathogenesis and immune escape is crucial for assessing the threat of new variants to public health. This review focuses on the emergence and pathogenesis of VOC, with particular emphasis on their evasion of neutralizing antibodies. Furthermore, the memory B cell, CD4+, and CD8+ T cell memory induced by different COVID-19 vaccines or infections are discussed, along with how these cells recognize VOC. This review summarizes the current knowledge on adaptive immunology regarding SARS-CoV-2 infection and vaccines. Such knowledge may also be applied to vaccine design for other pathogens.
Collapse
Affiliation(s)
- Changxu Chen
- Center for Infectious Disease Research, School of Life Science, Westlake University, Hangzhou, Zhejiang 310001, China
| | - Xin Wang
- Center for Infectious Disease Research, School of Life Science, Westlake University, Hangzhou, Zhejiang 310001, China
| | - Zeli Zhang
- Center for Infectious Disease Research, School of Life Science, Westlake University, Hangzhou, Zhejiang 310001, China.
| |
Collapse
|
32
|
Pitiriga VC, Papamentzelopoulou M, Konstantinakou KE, Vasileiou IV, Sakellariou KS, Spyrou NI, Tsakris A. Persistence of T-Cell Immunity Responses against SARS-CoV-2 for over 12 Months Post COVID-19 Infection in Unvaccinated Individuals with No Detectable IgG Antibodies. Vaccines (Basel) 2023; 11:1764. [PMID: 38140169 PMCID: PMC10747023 DOI: 10.3390/vaccines11121764] [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: 10/27/2023] [Revised: 11/09/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Immune response to SARS-CoV-2 is crucial for preventing reinfection or reducing disease severity. T-cells' long-term protection, elicited either by COVID-19 vaccines or natural infection, has been extensively studied thus far; however, it is still attracting considerable scientific interest. The aim of the present epidemiological study was to define the levels of T-cellular immunity response in a specific group of unvaccinated individuals from the general population with a prior confirmed COVID-19 infection and no measurable levels of IgG antibodies. METHODS We performed a retrospective descriptive analysis of data collected from the medical records of consecutive unvaccinated individuals recovered from COVID-19, who had proceeded to a large private medical center in the Attica region from September 2021 to September 2022 in order to be examined on their own initiative for SARS-CoV-2 T-cell immunity response. The analysis of T-cell responses was divided into three time periods post infection: Group A: up to 6 months; Group B: 6-12 months; Group C: >12 months. The SARS-CoV-2 T-cell response was estimated against spike (S) and nucleocapsid (N) structural proteins by performing the T-SPOT. COVID test methodology. SARS-CoV-2 IgG antibody levels were measured by the SARS-CoV-2 IgG II Quant assay (Abbott Diagnostics). RESULTS A total of 182 subjects were retrospectively included in the study, 85 females (46.7%) and 97 (53.3%) males, ranging from 19 to 91 years old (mean 50.84 ± 17.2 years). Among them, 59 (32.4%) had been infected within the previous 6 months from the examination date (Group A), 69 (37.9%) had been infected within a time period > 6 months and <1 year (Group B) and 54 (29.7%) had been infected within a time period longer than 1 year from the examination date (Group C). Among the three groups, a positive T-cell reaction against the S antigen was reported in 47/58 (81%) of Group A, 61/69 (88.4%) of Group B and 40/54 (74.1%) of Group C (chi square, p = 0.27). T-cell reaction against the N antigen was present in 45/58 (77.6%) of Group A, 61/69 (88.4%) of Group B and 36/54 (66.7%) of Group C (chi square, p = 0.02). The median Spot-Forming Cells (SFC) count for the S antigen was 18 (range from 0-160) in Group A, 19 (range from 0-130) in Group B and 17 (range from 0-160) in Group C (Kruskal-Wallis test, p = 0.11; pairwise comparisons: groups A-B, p = 0.95; groups A-C, p = 0.89; groups B-C, p = 0.11). The median SFCs count for the N antigen was 14.5 (ranging from 0 to 116) for Group A, 24 (ranging from 0-168) in Group B and 16 (ranging from 0-112) for Group C (Kruskal-Wallis test, p = 0.01; pairwise comparisons: groups A-B, p = 0.02; groups A-C, p = 0.97; groups B-C, p = 0.03). CONCLUSIONS Our data suggest that protective adaptive T-cellular immunity following natural infection by SARS-CoV-2 may persist for over 12 months, despite the undetectable humoral element.
Collapse
Affiliation(s)
- Vassiliki C. Pitiriga
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece;
| | - Myrto Papamentzelopoulou
- Molecular Biology Unit, 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Kanella E. Konstantinakou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Irene V. Vasileiou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Konstantina S. Sakellariou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Natalia I. Spyrou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece;
| |
Collapse
|
33
|
Wang L, Nicols A, Turtle L, Richter A, Duncan CJA, Dunachie SJ, Klenerman P, Payne RP. T cell immune memory after covid-19 and vaccination. BMJ MEDICINE 2023; 2:e000468. [PMID: 38027416 PMCID: PMC10668147 DOI: 10.1136/bmjmed-2022-000468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023]
Abstract
The T cell memory response is a crucial component of adaptive immunity responsible for limiting or preventing viral reinfection. T cell memory after infection with the SARS-CoV-2 virus or vaccination is broad, and spans multiple viral proteins and epitopes, about 20 in each individual. So far the T cell memory response is long lasting and provides a high level of cross reactivity and hence resistance to viral escape by variants of the SARS-CoV-2 virus, such as the omicron variant. All current vaccine regimens tested produce robust T cell memory responses, and heterologous regimens will probably enhance protective responses through increased breadth. T cell memory could have a major role in protecting against severe covid-19 disease through rapid viral clearance and early presentation of epitopes, and the presence of cross reactive T cells might enhance this protection. T cell memory is likely to provide ongoing protection against admission to hospital and death, and the development of a pan-coronovirus vaccine might future proof against new pandemic strains.
Collapse
Affiliation(s)
- Lulu Wang
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Alex Nicols
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Lance Turtle
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Tropical and Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Alex Richter
- Institute of Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK
| | - Christopher JA Duncan
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
- Department of Infection and Tropical Medicine, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Susanna J Dunachie
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University Faculty of Science, Bangkok, Thailand
| | - Paul Klenerman
- Oxford University Hospitals NHS Foundation Trust, Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, Oxfordshire, UK
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Rebecca P Payne
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
34
|
Onyango TB, Zhou F, Bredholt G, Brokstad KA, Lartey S, Mohn KGI, Özgümüs T, Kittang BR, Linchausen DW, Shafiani S, Elyanow R, Blomberg B, Langeland N, Cox RJ. SARS-CoV-2 specific immune responses in overweight and obese COVID-19 patients. Front Immunol 2023; 14:1287388. [PMID: 38022529 PMCID: PMC10653322 DOI: 10.3389/fimmu.2023.1287388] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is a known risk factor for severe respiratory tract infections. In this prospective study, we assessed the impact of being obese or overweight on longitudinal SARS-CoV-2 humoral and cellular responses up to 18 months after infection. 274 patients provided blood samples at regular time intervals up to 18 months including obese (BMI ≥30, n=32), overweight (BMI 25-29.9, n=103) and normal body weight (BMI 18.5-24.9, n=134) SARS-CoV-2 patients. We determined SARS-CoV-2 spike-specific IgG, IgA, IgM levels by ELISA and neutralising antibody titres by neutralisation assay. RBD- and spike-specific memory B cells were investigated by ELISpot, spike- and non-spike-specific IFN-γ, IL-2 and IFN-γ/IL-2 secreting T cells by FluoroSpot and T cell receptor (TCR) sequencing was performed. Higher BMI correlated with increased COVID-19 severity. Humoral and cellular responses were stronger in overweight and obese patients than normal weight patients and associated with higher spike-specific IgG binding titres relative to neutralising antibody titres. Linear regression models demonstrated that BMI, age and COVID-19 severity correlated independently with higher SARS-CoV-2 immune responses. We found an increased proportion of unique SARS-CoV-2 specific T cell clonotypes after infection in overweight and obese patients. COVID-19 vaccination boosted humoral and cellular responses irrespective of BMI, although stronger immune boosting was observed in normal weight patients. Overall, our results highlight more severe disease and an over-reactivity of the immune system in overweight and obese patients after SARS-CoV-2 infection, underscoring the importance of recognizing overweight/obese individuals as a risk group for prioritisation for COVID-19 vaccination.
Collapse
Affiliation(s)
| | - Fan Zhou
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Geir Bredholt
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Karl A. Brokstad
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Safety, Chemistry and Biomedical Laboratory Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Sarah Lartey
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kristin G.-I. Mohn
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Türküler Özgümüs
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | | | | | | | | | - Bjørn Blomberg
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- National Advisory Unit for Tropical Infectious Diseases, Haukeland University Hospital, Bergen, Norway
| | - Nina Langeland
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- National Advisory Unit for Tropical Infectious Diseases, Haukeland University Hospital, Bergen, Norway
| | - Rebecca Jane Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
35
|
Misra P, Garg PK, Awasthi A, Kant S, Rai SK, Ahmad M, Guleria R, Deori TJ, Mandal S, Jaiswal A, Gongal G, Vishwakarma S, Bairwa M, Kumar R, Haldar P, Binayke A. Cell-Mediated Immunity (CMI) for SARS-CoV-2 Infection Among the General Population of North India: A Cross-Sectional Analysis From a Sub-sample of a Large Sero-Epidemiological Study. Cureus 2023; 15:e48824. [PMID: 38106811 PMCID: PMC10722242 DOI: 10.7759/cureus.48824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
Background Cell-mediated immunity (CMI), or specifically T-cell-mediated immunity, is proven to remain largely preserved against the variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), including Omicron. The persistence of cell-mediated immune response in individuals longitudinally followed up for an extended period remains largely unelucidated. To address this, the current study was planned to study whether the effect of cell-mediated immunity persists after an extended period of convalescence or vaccination. Methods Whole blood specimens of 150 selected participants were collected and tested for Anti-SARS-CoV-2 Interferon-gamma (IFN-γ) response. Ex vivo SARS-CoV-2-specific interferon-gamma Enzyme-linked Immunospot (IFN-γ ELISpot) assay was carried out to determine the levels of virus-specific IFN-γ producing cells in individual samples. Findings Out of all the samples tested for anti-SARS-CoV-2 T-cell-mediated IFN-γ response, 78.4% of samples were positive. The median (interquartile range) spots forming units (SFU) per million levels of SARS-CoV-2-specific IFN-γ producing cells of the vaccinated and diagnosed participants was 336 (138-474) while those who were vaccinated but did not have the disease diagnosis was 18 (0-102); the difference between the groups was statistically significant. Since almost all the participants were vaccinated, a similar pattern of significance was observed when the diagnosed and the never-diagnosed participants were compared, irrespective of their vaccination status. Interpretations Cell-mediated immunity against SARS-CoV-2 persisted, irrespective of age and sex of the participant, for more than six months of previous exposure. Participants who had a history of diagnosed COVID-19 infection had better T-cell response compared to those who had never been diagnosed, in spite of being vaccinated.
Collapse
Affiliation(s)
- Puneet Misra
- Epidemiology and Public Health, Centre for Community Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Pramod K Garg
- Gastroenterology, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Amit Awasthi
- Allergy and Immunology, Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, IND
| | - Shashi Kant
- Epidemiology and Public Health, Centre for Community Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Sanjay K Rai
- Epidemiology and Public Health, Centre for Community Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Mohammad Ahmad
- Epidemiology and Public Health, World Health Organization, New Delhi, IND
| | - Randeep Guleria
- Pulmonary, Critical Care, and Sleep Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Trideep J Deori
- Epidemiology and Public Health, Centre for Community Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Suprakash Mandal
- Epidemiology and Public Health, Centre for Community Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Abhishek Jaiswal
- Community Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Gaurav Gongal
- Epidemiology and Public Health, Centre for Community Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Siddhesh Vishwakarma
- Allergy and Immunology, Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, IND
| | - Mohan Bairwa
- Epidemiology and Public Health, Centre for Community Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Rakesh Kumar
- Epidemiology and Public Health, Centre for Community Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Partha Haldar
- Preventive Medicine, Centre for Community Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Akshay Binayke
- Allergy and Immunology, Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, IND
| |
Collapse
|
36
|
Ssali I, Mugaba S, Watelo AK, Bemanzi J, Katende JS, Oluka GK, Ankunda V, Baine C, Kato L, Onyachi N, Muwanga M, Jjuuko M, Kayiwa J, Nsereko C, Auma BO, Weiskopf D, Sette A, Lutalo T, Musenero M, Kaleebu P, Serwanga J. Spike protein is a key target for stronger and more persistent T-cell responses-a study of mild and asymptomatic SARS-CoV-2 infection. Int J Infect Dis 2023; 136:49-56. [PMID: 37683720 DOI: 10.1016/j.ijid.2023.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/08/2023] [Accepted: 09/02/2023] [Indexed: 09/10/2023] Open
Abstract
OBJECTIVES Understanding the immune response in very mild and asymptomatic COVID-19 is crucial for developing effective vaccines and immunotherapies, yet remains poorly characterized. This longitudinal study examined the evolution of interferon (IFN)-γ responses to SARS-CoV-2 peptides in 109 asymptomatic or mildly symptomatic Ugandan COVID-19 patients across 365 days and explored their association with antibody generation. METHODS T-cell responses to spike-containing clusters of differentiation (CD4)-S and CD8 nCoV-A (CD8-A) megapools, and the non-spike CD4-R and CD8 nCoV-B (CD8-B) megapools, were assessed and correlated with demographic and temporal variables. RESULTS SARS-CoV-2-specific IFN-γ responses were consistently detected in all peptide pools and time points, with the spike-targeted response exhibiting higher potency and durability than the non-spike responses. Throughout the entire 365-day infection timeline, a robust positive correlation was observed between CD4 T-cell responses to the spike-derived peptides and anti-spike immunoglobulin G antibody levels, underscoring their interdependent dynamics in the immune response against SARS-CoV-2; in contrast, CD8 T-cell responses exhibited no such correlation, highlighting their distinctive, autonomous role in defense. No meaningful variations in complete blood count parameters were observed between individuals with COVID-19 infection and those without, indicating clinical insignificance. CONCLUSIONS This study highlights the dominant role of spike-directed T-cell responses in mild and asymptomatic disease and provides crucial longitudinal data from Sub-Saharan African settings. The findings provide valuable insights into the dynamics of T-cell responses and their potential significance in developing effective strategies for combating COVID-19.
Collapse
Affiliation(s)
- Ivan Ssali
- Pathogen Genomics, Phenotype, and Immunity Program, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Susan Mugaba
- Pathogen Genomics, Phenotype, and Immunity Program, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | | | - Juliana Bemanzi
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Joseph Ssebwana Katende
- Pathogen Genomics, Phenotype, and Immunity Program, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda; Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Gerald Kevin Oluka
- Pathogen Genomics, Phenotype, and Immunity Program, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda; Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Violet Ankunda
- Pathogen Genomics, Phenotype, and Immunity Program, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Claire Baine
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Laban Kato
- Pathogen Genomics, Phenotype, and Immunity Program, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Nathan Onyachi
- Department of Internal Medicine, Masaka Regional Referral Hospital, Masaka, Uganda
| | - Moses Muwanga
- Department of Internal Medicine, Entebbe Regional Referral Hospital, Entebbe, Uganda
| | - Mark Jjuuko
- Department of Internal Medicine, Masaka Regional Referral Hospital, Masaka, Uganda
| | - John Kayiwa
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Christopher Nsereko
- Department of Internal Medicine, Entebbe Regional Referral Hospital, Entebbe, Uganda
| | - Betty Oliver Auma
- Pathogen Genomics, Phenotype, and Immunity Program, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, USA
| | - Tom Lutalo
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Monica Musenero
- Science, Technology, and Innovation Secretariat, Office of the President, Government of Uganda, Kampala, Uganda
| | - Pontiano Kaleebu
- Pathogen Genomics, Phenotype, and Immunity Program, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda; Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Jennifer Serwanga
- Pathogen Genomics, Phenotype, and Immunity Program, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda; Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda.
| |
Collapse
|
37
|
Maurya DK, Sharma D. Culinary spices and herbs in managing early and long-COVID-19 complications: A comprehensive review. Phytother Res 2023; 37:4908-4931. [PMID: 37468320 DOI: 10.1002/ptr.7957] [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: 12/22/2022] [Revised: 06/16/2023] [Accepted: 07/02/2023] [Indexed: 07/21/2023]
Abstract
Human race is preparing for the upsurge and aftermath of COVID-19 pandemic complicated by novel variants, new waves, variable mortality rate, and post-COVID complications. Despite use of repurposed drugs, symptomatic treatments and licensing of multiple vaccines, the daily number of cases and rate of transmission are significant. Culinary spices and herbs have been historically used in pandemic and non-pandemic times to reduce respiratory viral burden. Specific food items and culinary spices can boost the levels of protective immunity and also offer therapeutic benefits against impervious bugs via well-known as well as less-known but scientifically testable mechanisms. Here, we analyzed the phytochemicals profile of Ayurvedic herbs and inferred from the clinical trials/observational studies to provide a focused and succinct perspective on the relevance of "food-based" traditional decoction to moderate COVID-19 disease and long-COVID via modulation of immunity and reinstatement of homeostasis. We also underscore the druggable targets in pathogenesis of COVID-19 which are relevant to the ongoing clinical trials using spices and herbs. This information will provide a strong scientific rationale for standardization of the traditional herbs-based therapies and adopting the use of herbs, spices, and their formulations for reducing SARS-CoV-2 transmission, long-COVID symptoms, and COVID-19 disease progression.
Collapse
Affiliation(s)
- Dharmendra Kumar Maurya
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
38
|
Mohan A, Iyer VA, Kumar D, Batra L, Dahiya P. Navigating the Post-COVID-19 Immunological Era: Understanding Long COVID-19 and Immune Response. Life (Basel) 2023; 13:2121. [PMID: 38004261 PMCID: PMC10672162 DOI: 10.3390/life13112121] [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: 07/06/2023] [Revised: 09/25/2023] [Accepted: 09/30/2023] [Indexed: 11/26/2023] Open
Abstract
The COVID-19 pandemic has affected the world unprecedentedly, with both positive and negative impacts. COVID-19 significantly impacted the immune system, and understanding the immunological consequences of COVID-19 is essential for developing effective treatment strategies. The purpose of this review is to comprehensively explore and provide insights into the immunological aspects of long COVID-19, a phenomenon where individuals continue to experience a range of symptoms and complications, even after the acute phase of COVID-19 infection has subsided. The immune system responds to the initial infection by producing various immune cells and molecules, including antibodies, T cells, and cytokines. However, in some patients, this immune response becomes dysregulated, leading to chronic inflammation and persistent symptoms. Long COVID-19 encompasses diverse persistent symptoms affecting multiple organ systems, including the respiratory, cardiovascular, neurological, and gastrointestinal systems. In the post-COVID-19 immunological era, long COVID-19 and its impact on immune response have become a significant concern. Post-COVID-19 immune pathology, including autoimmunity and immune-mediated disorders, has also been reported in some patients. This review provides an overview of the current understanding of long COVID-19, its relationship to immunological responses, and the impact of post-COVID-19 immune pathology on patient outcomes. Additionally, the review addresses the current and potential treatments for long COVID-19, including immunomodulatory therapies, rehabilitation programs, and mental health support, all of which aim to improve the quality of life for individuals with long COVID-19. Understanding the complex interplay between the immune system and long COVID-19 is crucial for developing targeted therapeutic strategies and providing optimal care in the post-COVID-19 era.
Collapse
Affiliation(s)
- Aditi Mohan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida Sector-125, Noida 201313, Uttar Pradesh, India; (A.M.); (V.A.I.)
| | - Venkatesh Anand Iyer
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida Sector-125, Noida 201313, Uttar Pradesh, India; (A.M.); (V.A.I.)
| | - Dharmender Kumar
- Department of Biotechnology, Deenbandhu Chhotu Ram University of Science &Technology, Murthal, Sonipat 131309, Haryana, India;
| | - Lalit Batra
- Regional Biocontainment Laboratory, Center for Predictive Medicine, University of Louisville, Louisville, KY 40222, USA
| | - Praveen Dahiya
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida Sector-125, Noida 201313, Uttar Pradesh, India; (A.M.); (V.A.I.)
| |
Collapse
|
39
|
Offor UT, Hollis P, Ognjanovic M, Parry G, Khushnood A, Long HM, Gennery AR, Bacon CM, Simmonds J, Reinhardt Z, Bomken S. Immunology of THymectomy And childhood CArdiac transplant (ITHACA): protocol for a UK-wide prospective observational cohort study to identify immunological risk factors of post-transplant lymphoproliferative disease (PTLD) in thymectomised children. BMJ Open 2023; 13:e079582. [PMID: 37865406 PMCID: PMC10603490 DOI: 10.1136/bmjopen-2023-079582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/10/2023] [Indexed: 10/23/2023] Open
Abstract
INTRODUCTION Paediatric heart transplant patients are disproportionately affected by Epstein-Barr virus (EBV)-related post-transplant lymphoproliferative disease (PTLD) compared with other childhood solid organ recipients. The drivers for this disparity remain poorly understood. A potential risk factor within this cohort is the routine surgical removal of the thymus-a gland critical for the normal development of T-lymphocyte-mediated antiviral immunity-in early life, which does not occur in other solid organ transplant recipients. Our study aims to describe the key immunological differences associated with early thymectomy, its impact on the temporal immune response to EBV infection and subsequent risk of PTLD. METHODS AND ANALYSIS Prospective and sequential immune monitoring will be performed for 34 heart transplant recipients and 6 renal transplant patients (aged 0-18 years), stratified into early (<1 year), late (>1 year) and non-thymectomy groups. Peripheral blood samples and clinical data will be taken before transplant and at 3, 6, 12 and 24 months post-transplant. Single cell analysis of circulating immune cells and enumeration of EBV-specific T-lymphocytes will be performed using high-dimensional spectral flow cytometry with peptide-Major Histocompatibilty Complex (pMHC) I/II tetramer assay, respectively. The functional status of EBV-specific T-lymphocytes, along with EBV antibodies and viral load will be monitored at each of the predefined study time points. ETHICS AND DISSEMINATION Ethical approval for this study has been obtained from the North of Scotland Research Ethics Committee. The results will be disseminated through publications in peer-reviewed journals, presentations at scientific conferences and patient-centred forums, including social media. TRIAL REGISTRATION NUMBER ISRCTN10096625.
Collapse
Affiliation(s)
- Ugonna T Offor
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Department of Paediatric Haematology and Oncology, Great North Children's Hospital, Newcastle Upon Tyne, UK
| | - Paolo Hollis
- Department of Cardiothoracic Transplant, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Milos Ognjanovic
- Department of Paediatric Nephrology, Great North Children's Hospital, Newcastle Upon Tyne, UK
| | - Gareth Parry
- Department of Cardiopulmonary Transplantation, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Abbas Khushnood
- Department of Cardiopulmonary Transplantation, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Heather M Long
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Andrew R Gennery
- Translational and Clinical Research Institute, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Department of Paediatric Immunology and Haematopoietic Stem Cell Transplantation, Great North Children's Hospital, Newcastle Upon Tyne, UK
| | - Chris M Bacon
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Department of Cellular Pathology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Jacob Simmonds
- Department of Cardiothoracic Transplant, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Zdenka Reinhardt
- Department of Cardiopulmonary Transplantation, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Simon Bomken
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Department of Paediatric Haematology and Oncology, Great North Children's Hospital, Newcastle Upon Tyne, UK
| |
Collapse
|
40
|
Khadri L, Ziraksaz MH, Barekzai AB, Ghauri B. T cell responses to SARS-CoV-2. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 202:183-217. [PMID: 38237986 DOI: 10.1016/bs.pmbts.2023.06.001] [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: 01/23/2024]
Abstract
This chapter provides a comprehensive analysis of T cell responses in COVID-19, focusing on T cell differentiation, specificity, and functional characteristics during SARS-CoV-2 infection. The differentiation of T cells in COVID-19 is explored, highlighting the key factors that influence T cell fate and effector functions. The immunology of the spike protein, a critical component of SARS-CoV-2, is discussed in detail, emphasizing its role in driving T-cell responses. The cellular immune responses against SARS-CoV-2 during acute infection are examined, including the specificity, phenotype, and functional attributes of SARS-CoV-2-specific T-cell responses. Furthermore, the chapter explores T-cell cross-recognition against other human coronaviruses (HCoVs) and the mechanisms of immune regulation mediated by spike proteins. This includes the induction of regulation through the innate immune system, the activation of self-spike protein-cross-reactive regulatory T cells, and the impact of self-tolerance on the regulation of spike proteins. The chapter investigates T cell responses to self-spike proteins and their implications in disease. The role of spike proteins as immunological targets in the context of COVID-19 is examined, shedding light on potential therapeutic interventions and clinical trials in autoimmune diseases. In conclusion, this chapter provides a comprehensive understanding of T cell responses in COVID-19, highlighting their differentiation, immune regulation, and clinical implications. This knowledge contributes to the development of targeted immunotherapies, vaccine strategies, and diagnostic approaches for COVID-19 and other related diseases.
Collapse
Affiliation(s)
- Laiqha Khadri
- Department of Biotechnology, Immune Inspired, Bangalore.
| | | | | | - Baber Ghauri
- Department of Biotechnology, Immune Inspired, Bangalore
| |
Collapse
|
41
|
Groß-Albenhausen E, Weier A, Velten M, Heider T, Chunder R, Kuerten S. Immune monitoring of SARS-CoV-2-specific T cell and B cell responses in patients with multiple sclerosis treated with ocrelizumab. Front Immunol 2023; 14:1254128. [PMID: 37841269 PMCID: PMC10569464 DOI: 10.3389/fimmu.2023.1254128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/30/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction Since the development of the coronavirus disease (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), there has been significant interest in determining the effectiveness of SARS-CoV-2 vaccines in patients under immunomodulatory or immunosuppressive therapies. The aim of this study was to evaluate the impact of ocrelizumab, a monoclonal anti-CD20 antibody, on SARS-CoV-2-specific T cell and B cell responses in patients with relapsing-remitting multiple sclerosis (RRMS). Methods To this end, peripheral blood mononuclear cells (PBMCs) were isolated from n = 23 patients with RRMS. Of these patients, n = 17 were tested before (time point t0) and one month after (time point t1) their first dose of ocrelizumab. In addition, we studied n = 9 RRMS patients that got infected with SARS-CoV-2 over the course of ocrelizumab therapy (time point t2). PBMCs were also isolated from n = 19 age- and gender-matched healthy controls (HCs) after vaccination or infection with SARS-CoV-2, respectively. Interferon-γ (IFN-γ)/interleukin-2 (IL-2) and granzyme B (GzB)/perforin (PFN) double-color enzyme-linked immunospot (ELISPOT) assays or single-color ELISPOT assays were performed to measure SARS-CoV-2 antigen-specific T cell and B cell responses. Anti-viral antibody titers were quantified in the serum by chemiluminescence immunoassay. Results Our data indicate a significant difference in the SARS-CoV-2 specific IFN-γ (P = 0.0119) and PFN (P = 0.0005) secreting T cell compartment in the MS cohort at t0 compared to HCs. Following the first dose of ocrelizumab treatment, a significant decrease in the number of SARS-CoV-2 spike protein-specific B cells was observed (P = 0.0012). Infection with SARS-CoV-2 in MS patients under ocrelizumab therapy did not significantly alter their existing immune response against the virus. Kaplan-Meier survival analysis suggested that the spike S1 protein-specific immunoglobulin (Ig)G response might be a key parameter for predicting the probability of (re)infection with SARS-CoV-2. Discussion Our results call for a critical discussion regarding appropriate vaccination intervals and potential biomarkers for the prediction of (re)infection with SARS-CoV-2 in patients with MS receiving ocrelizumab. Unique identifier DRKS00029110; URL: http://apps.who.int/trialsearch/.
Collapse
Affiliation(s)
- Elina Groß-Albenhausen
- Institute of Neuroanatomy, Faculty of Medicine, University of Bonn and University Hospital Bonn, Bonn, Germany
| | - Alicia Weier
- Institute of Neuroanatomy, Faculty of Medicine, University of Bonn and University Hospital Bonn, Bonn, Germany
| | - Markus Velten
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Bonn, Bonn, Germany
| | - Thorsten Heider
- Clinic for Neurology, Klinikum St. Marien Amberg, Amberg, Germany
| | - Rittika Chunder
- Institute of Neuroanatomy, Faculty of Medicine, University of Bonn and University Hospital Bonn, Bonn, Germany
| | - Stefanie Kuerten
- Institute of Neuroanatomy, Faculty of Medicine, University of Bonn and University Hospital Bonn, Bonn, Germany
| |
Collapse
|
42
|
Kiselev IN, Akberdin IR, Kolpakov FA. Delay-differential SEIR modeling for improved modelling of infection dynamics. Sci Rep 2023; 13:13439. [PMID: 37596296 PMCID: PMC10439236 DOI: 10.1038/s41598-023-40008-9] [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: 06/23/2022] [Accepted: 08/03/2023] [Indexed: 08/20/2023] Open
Abstract
SEIR (Susceptible-Exposed-Infected-Recovered) approach is a classic modeling method that is frequently used to study infectious diseases. However, in the vast majority of such models transitions from one population group to another are described using the mass-action law. That causes inability to reproduce observable dynamics of an infection such as the incubation period or progression of the disease's symptoms. In this paper, we propose a new approach to simulate the epidemic dynamics based on a system of differential equations with time delays and instant transitions to approximate durations of transition processes more correctly and make model parameters more clear. The suggested approach can be applied not only to Covid-19 but also to the study of other infectious diseases. We utilized it in the development of the delay-based model of the COVID-19 pandemic in Germany and France. The model takes into account testing of different population groups, symptoms progression from mild to critical, vaccination, duration of protective immunity and new virus strains. The stringency index was used as a generalized characteristic of the non-pharmaceutical government interventions in corresponding countries to contain the virus spread. The parameter identifiability analysis demonstrated that the presented modeling approach enables to significantly reduce the number of parameters and make them more identifiable. Both models are publicly available.
Collapse
Affiliation(s)
- I N Kiselev
- FRC for Information and Computational Technologies, Novosibirsk, Russia.
- Sirius University of Science and Technology, Sirius, Russia.
- BIOSOFT.RU, Ltd, Novosibirsk, Russia.
| | - I R Akberdin
- Sirius University of Science and Technology, Sirius, Russia
- BIOSOFT.RU, Ltd, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - F A Kolpakov
- FRC for Information and Computational Technologies, Novosibirsk, Russia
- Sirius University of Science and Technology, Sirius, Russia
- BIOSOFT.RU, Ltd, Novosibirsk, Russia
| |
Collapse
|
43
|
Dang TTT, Anzurez A, Nakayama-Hosoya K, Miki S, Yamashita K, de Souza M, Matano T, Kawana-Tachikawa A. Breadth and Durability of SARS-CoV-2-Specific T Cell Responses following Long-Term Recovery from COVID-19. Microbiol Spectr 2023; 11:e0214323. [PMID: 37428088 PMCID: PMC10433967 DOI: 10.1128/spectrum.02143-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
T cell immunity is crucial for long-term immunological memory, but the profile of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific memory T cells in individuals who recovered from COVID-19 (COVID-19-convalescent individuals) is not sufficiently assessed. In this study, the breadth and magnitude of SARS-CoV-2-specific T cell responses were determined in COVID-19-convalescent individuals in Japan. Memory T cells against SARS-CoV-2 were detected in all convalescent individuals, and those with more severe disease exhibited a broader T cell response relative to cases with mild symptoms. Comprehensive screening of T cell responses at the peptide level was conducted for spike (S) and nucleocapsid (N) proteins, and regions frequently targeted by T cells were identified. Multiple regions in S and N proteins were targeted by memory T cells, with median numbers of target regions of 13 and 4, respectively. A maximum of 47 regions were recognized by memory T cells for an individual. These data indicate that SARS-CoV-2-convalescent individuals maintain a substantial breadth of memory T cells for at least several months following infection. Broader SARS-CoV-2-specific CD4+ T cell responses, relative to CD8+ T cell responses, were observed for the S but not the N protein, suggesting that antigen presentation is different between viral proteins. The binding affinity of predicted CD8+ T cell epitopes to HLA class I molecules in these regions was preserved for the Delta variant and at 94 to 96% for SARS-CoV-2 Omicron subvariants, suggesting that the amino acid changes in these variants do not have a major impact on antigen presentation to SARS-CoV-2-specific CD8+ T cells. IMPORTANCE RNA viruses, including SARS-CoV-2, evade host immune responses through mutations. As broader T cell responses against multiple viral proteins could minimize the impact of each single amino acid mutation, the breadth of memory T cells would be one essential parameter for effective protection. In this study, breadth of memory T cells to S and N proteins was assessed in COVID-19-convalescent individuals. While broad T cell responses were induced against both proteins, the ratio of N to S proteins for breadth of T cell responses was significantly higher in milder cases. The breadth of CD4+ and CD8+ T cell responses was also significantly different between S and N proteins, suggesting different contributions of N and S protein-specific T cells for COVID-19 control. Most CD8+ T cell epitopes in the immunodominant regions maintained their HLA binding to SARS-CoV-2 Omicron subvariants. Our study provides insights into understanding the protective efficacy of SARS-CoV-2-specific memory T cells against reinfection.
Collapse
Affiliation(s)
- Thi Thu Thao Dang
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Alitzel Anzurez
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | | | - Shoji Miki
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | | | - Mark de Souza
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
- Department of AIDS Vaccine Development, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Ai Kawana-Tachikawa
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
- Department of AIDS Vaccine Development, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
44
|
Akanmu S, Herrera BB, Chaplin B, Ogunsola S, Osibogun A, Onawoga F, John-Olabode S, Akase IE, Nwosu A, Hamel DJ, Chang CA, Kanki PJ. High SARS-CoV-2 seroprevalence in Lagos, Nigeria with robust antibody and cellular immune responses. JOURNAL OF CLINICAL VIROLOGY PLUS 2023; 3:100156. [PMID: 37388808 PMCID: PMC10289822 DOI: 10.1016/j.jcvp.2023.100156] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/01/2023] Open
Abstract
Background Early evidence suggested that the impact of the COVID-19 pandemic was less severe in Africa compared to other parts of the world. However, more recent studies indicate higher SARS-CoV-2 infection and COVID-19 mortality rates on the continent than previously documented. Research is needed to better understand SARS-CoV-2 infection and immunity in Africa. Methods In early 2021, we studied the immune responses in healthcare workers (HCWs) at Lagos University Teaching Hospital (n = 134) and Oxford-AstraZeneca COVID-19 vaccine recipients from the general population (n = 116) across five local government areas (LGAs) in Lagos State, Nigeria. Western blots were used to simultaneously detect SARS-CoV-2 spike and nucleocapsid (N) antibodies (n = 250), and stimulation of peripheral blood mononuclear cells with N followed by an IFN-γ ELISA was used to examine T cell responses (n = 114). Results Antibody data demonstrated high SARS-CoV-2 seroprevalence of 72·4% (97/134) in HCWs and 60·3% (70/116) in the general population. Antibodies directed to only SARS-CoV-2 N, suggesting pre-existing coronavirus immunity, were seen in 9·7% (13/134) of HCWs and 15·5% (18/116) of the general population. T cell responses against SARS-CoV-2 N (n = 114) were robust in detecting exposure to the virus, demonstrating 87·5% sensitivity and 92·9% specificity in a subset of control samples tested. T cell responses against SARS-CoV-2 N were also observed in 83.3% of individuals with N-only antibodies, further suggesting that prior non-SARS-CoV-2 coronavirus infection may provide cellular immunity to SARS-CoV-2. Conclusions These results have important implications for understanding the paradoxically high SARS-CoV-2 infection with low mortality rate in Africa and supports the need to better understand the implications of SARS-CoV-2 cellular immunity.
Collapse
Affiliation(s)
- Sulaimon Akanmu
- Lagos University Teaching Hospital, College of Medicine, University of Lagos, Eyo Ita Crescent, Akoka Yaba, Lagos, Nigeria
| | - Bobby Brooke Herrera
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 651 Huntington Avenue, Boston, MA, USA
- Department of Medicine, Division of Allergy, Immunology, and Infectious Diseases, and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, 89 French St, New Brunswick, NJ, USA
- Rutgers Global Health Institute, Rutgers University, 112 Paterson Street, New Brunswick, NJ, USA
- Mir Biosciences, Inc., 12 Depot Way, Dunellen, NJ, USA
| | - Beth Chaplin
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 651 Huntington Avenue, Boston, MA, USA
| | - Sade Ogunsola
- Lagos University Teaching Hospital, College of Medicine, University of Lagos, Eyo Ita Crescent, Akoka Yaba, Lagos, Nigeria
| | - Akin Osibogun
- Lagos University Teaching Hospital, College of Medicine, University of Lagos, Eyo Ita Crescent, Akoka Yaba, Lagos, Nigeria
- Lagos State COVID-19 Taskforce, Lagos State Ministry of Health, Lagos, Nigeria
| | - Fatima Onawoga
- Lagos University Teaching Hospital, College of Medicine, University of Lagos, Eyo Ita Crescent, Akoka Yaba, Lagos, Nigeria
| | - Sarah John-Olabode
- Lagos University Teaching Hospital, College of Medicine, University of Lagos, Eyo Ita Crescent, Akoka Yaba, Lagos, Nigeria
| | - Iorhen E Akase
- Lagos University Teaching Hospital, College of Medicine, University of Lagos, Eyo Ita Crescent, Akoka Yaba, Lagos, Nigeria
| | - Augustina Nwosu
- Lagos University Teaching Hospital, College of Medicine, University of Lagos, Eyo Ita Crescent, Akoka Yaba, Lagos, Nigeria
| | - Donald J Hamel
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 651 Huntington Avenue, Boston, MA, USA
| | - Charlotte A Chang
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 651 Huntington Avenue, Boston, MA, USA
| | - Phyllis J Kanki
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 651 Huntington Avenue, Boston, MA, USA
| |
Collapse
|
45
|
Fallet B, Foglierini M, Porret R, Alcaraz-Serna A, Sauvage C, Jenelten R, Caplanusi T, Gilliet M, Perez L, Fenwick C, Genolet R, Harari A, Bobisse S, Gottardo R, Pantaleo G, Muller YD. Intradermal skin test with mRNA vaccines as a surrogate marker of T cell immunity in immunocompromised patients. J Infect 2023; 87:111-119. [PMID: 37321353 PMCID: PMC10264165 DOI: 10.1016/j.jinf.2023.06.005] [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: 04/02/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023]
Abstract
OBJECTIVES Intradermal skin test (IDT) with mRNA vaccines may represent a simple, reliable, and affordable tool to measure T cell response in immunocompromised patients who failed to mount serological responses following vaccination with mRNA covid-19 vaccines. METHODS We compared anti-SARS-CoV-2 antibodies and cellular responses in vaccinated immunocompromised patients (n = 58), healthy seronegative naive controls (NC, n = 8), and healthy seropositive vaccinated controls (VC, n = 32) by Luminex, spike-induced IFN-γ Elispot and an IDT. A skin biopsy 24 h after IDT and single-cell RNAseq was performed in three vaccinated volunteers. RESULTS Twenty-five percent of seronegative NC had a positive Elispot (2/8) and IDT (1/4), compared to 95% (20/21) and 93% (28/30) in seropositive VC, respectively. Single-cell RNAseq data in the skin of VC showed a predominant mixed population of effector helper and cytotoxic T cells. The TCR repertoire revealed 18/1064 clonotypes with known specificities against SARS-CoV-2, among which six were spike-specific. Seronegative immunocompromised patients with positive Elispot and IDT were in 83% (5/6) treated with B cell-depleting reagents, while those with negative IDT were all transplant recipients. CONCLUSIONS Our results indicate that delayed local reaction to IDT reflects vaccine-induced T-cell immunity opening new perspectives to monitor seronegative patients and elderly populations with waning immunity.
Collapse
Affiliation(s)
- Benedict Fallet
- Division of Immunology and Allergy, University Hospital of Lausanne, Switzerland
| | - Mathilde Foglierini
- Division of Immunology and Allergy, University Hospital of Lausanne, Switzerland; Biomedical Data Science Center, University Hospital of Lausanne, Switzerland
| | - Raphael Porret
- Division of Immunology and Allergy, University Hospital of Lausanne, Switzerland
| | - Ana Alcaraz-Serna
- Division of Immunology and Allergy, University Hospital of Lausanne, Switzerland
| | - Christophe Sauvage
- Center for Cell Immunotherapy, Department of Oncology and Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne CH-1066, Switzerland
| | - Raphael Jenelten
- Department of Dermatology and Venereology, University Hospital of Lausanne, Switzerland
| | - Teofila Caplanusi
- Department of Dermatology and Venereology, University Hospital of Lausanne, Switzerland
| | - Michel Gilliet
- Department of Dermatology and Venereology, University Hospital of Lausanne, Switzerland; University of Lausanne, Faculty of Biology and Medicine, Lausanne, Switzerland
| | - Laurent Perez
- Division of Immunology and Allergy, University Hospital of Lausanne, Switzerland; University of Lausanne, Faculty of Biology and Medicine, Lausanne, Switzerland
| | - Craig Fenwick
- Division of Immunology and Allergy, University Hospital of Lausanne, Switzerland
| | - Raphael Genolet
- Center for Cell Immunotherapy, Department of Oncology and Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne CH-1066, Switzerland
| | - Alexandre Harari
- Center for Cell Immunotherapy, Department of Oncology and Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne CH-1066, Switzerland; University of Lausanne, Faculty of Biology and Medicine, Lausanne, Switzerland
| | - Sara Bobisse
- Center for Cell Immunotherapy, Department of Oncology and Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne CH-1066, Switzerland
| | - Raphael Gottardo
- Biomedical Data Science Center, University Hospital of Lausanne, Switzerland; University of Lausanne, Faculty of Biology and Medicine, Lausanne, Switzerland; Swiss Institute for Bioinformatics, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Division of Immunology and Allergy, University Hospital of Lausanne, Switzerland; University of Lausanne, Faculty of Biology and Medicine, Lausanne, Switzerland
| | - Yannick D Muller
- Division of Immunology and Allergy, University Hospital of Lausanne, Switzerland; University of Lausanne, Faculty of Biology and Medicine, Lausanne, Switzerland.
| |
Collapse
|
46
|
Abdelaziz MO, Raftery MJ, Weihs J, Bielawski O, Edel R, Köppke J, Vladimirova D, Adler JM, Firsching T, Voß A, Gruber AD, Hummel LV, Fernandez Munoz I, Müller-Marquardt F, Willimsky G, Elleboudy NS, Trimpert J, Schönrich G. Early protective effect of a ("pan") coronavirus vaccine (PanCoVac) in Roborovski dwarf hamsters after single-low dose intranasal administration. Front Immunol 2023; 14:1166765. [PMID: 37520530 PMCID: PMC10372429 DOI: 10.3389/fimmu.2023.1166765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has highlighted the danger posed by human coronaviruses. Rapid emergence of immunoevasive variants and waning antiviral immunity decrease the effect of the currently available vaccines, which aim at induction of neutralizing antibodies. In contrast, T cells are marginally affected by antigen evolution although they represent the major mediators of virus control and vaccine protection against virus-induced disease. Materials and methods We generated a multi-epitope vaccine (PanCoVac) that encodes the conserved T cell epitopes from all structural proteins of coronaviruses. PanCoVac contains elements that facilitate efficient processing and presentation of PanCoVac-encoded T cell epitopes and can be uploaded to any available vaccine platform. For proof of principle, we cloned PanCoVac into a non-integrating lentivirus vector (NILV-PanCoVac). We chose Roborovski dwarf hamsters for a first step in evaluating PanCoVac in vivo. Unlike mice, they are naturally susceptible to SARS-CoV-2 infection. Moreover, Roborovski dwarf hamsters develop COVID-19-like disease after infection with SARS-CoV-2 enabling us to look at pathology and clinical symptoms. Results Using HLA-A*0201-restricted reporter T cells and U251 cells expressing a tagged version of PanCoVac, we confirmed in vitro that PanCoVac is processed and presented by HLA-A*0201. As mucosal immunity in the respiratory tract is crucial for protection against respiratory viruses such as SARS-CoV-2, we tested the protective effect of single-low dose of NILV-PanCoVac administered via the intranasal (i.n.) route in the Roborovski dwarf hamster model of COVID-19. After infection with ancestral SARS-CoV-2, animals immunized with a single-low dose of NILV-PanCoVac i.n. did not show symptoms and had significantly decreased viral loads in the lung tissue. This protective effect was observed in the early phase (2 days post infection) after challenge and was not dependent on neutralizing antibodies. Conclusion PanCoVac, a multi-epitope vaccine covering conserved T cell epitopes from all structural proteins of coronaviruses, might protect from severe disease caused by SARS-CoV-2 variants and future pathogenic coronaviruses. The use of (HLA-) humanized animal models will allow for further efficacy studies of PanCoVac-based vaccines in vivo.
Collapse
Affiliation(s)
- Mohammed O. Abdelaziz
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Martin J. Raftery
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julian Weihs
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Pediatrics, Division of Gastroenterology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Olivia Bielawski
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Richard Edel
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julia Köppke
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Julia M. Adler
- Institute of Virology, Freie Universität Berlin, Berlin, Germany
| | - Theresa Firsching
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Anne Voß
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Achim D. Gruber
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Luca V. Hummel
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ivan Fernandez Munoz
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Francesca Müller-Marquardt
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gerald Willimsky
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Partner Site Berlin, Berlin, Germany
| | - Nooran S. Elleboudy
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Jakob Trimpert
- Institute of Virology, Freie Universität Berlin, Berlin, Germany
| | - Günther Schönrich
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
47
|
Bensaid K, Lamara Mahammed L, Habchi K, Saidani M, Allam I, Djidjik R. Evaluation of the Humoral and Cellular Immune Response Post COVID-19 Infection in Kidney Transplant Recipients. J Clin Med 2023; 12:3900. [PMID: 37373595 DOI: 10.3390/jcm12123900] [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: 04/28/2023] [Revised: 05/21/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Kidney transplantation is a major risk factor for severe forms of coronavirus disease 2019 (COVID-19). The dynamics and the persistence of the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in this immunocompromised population remain largely unknown. This study aimed to evaluate the persistence of humoral and cellular immune response in kidney transplant recipients (KTRs) and to establish whether immunosuppressive therapy influenced long-term immunity in this population. We report here the analysis of anti-SARS-CoV-2 antibodies and T cell-mediated immune responses in 36 KTRs compared to a control group who recovered from mild COVID-19. After a mean time of 5.22 ± 0.96 months post symptom onset for kidney transplant recipients, 97.22% of patients and 100% of the control group displayed anti-S1 immunoglobulin G SARS-CoV-2 antibodies (p > 0.05). No significant difference was reported in the median of neutralizing antibodies between the groups (97.50 [55.25-99] in KTRs vs. 84 [60-98] in control group, p = 0.35). A significant difference in SARS-CoV-2-specific T cell reactivity was found in the KTRs compared to the healthy controls. The levels of IFNγ release after stimulation by Ag1, Ag2 and Ag3 were higher in the control group compared to the kidney transplant group (p = 0.007, p = 0.025 and p = 0.008, respectively). No statistically significant correlation between humoral and cellular immunity was found in the KTRs. Our findings indicated that humoral immunity persisted similarly for up to 4 to 6 months post symptom onset in both the KTRs and the control group; however, T cell response was significantly higher in the healthy population compared to the immunocompromised patients.
Collapse
Affiliation(s)
- Kahina Bensaid
- Immunology Department, Beni-Messous Teaching Hospital, Faculty of Pharmacy, University of Algiers, Algiers 16000, Algeria
| | - Lydia Lamara Mahammed
- Immunology Department, Beni-Messous Teaching Hospital, Faculty of Pharmacy, University of Algiers, Algiers 16000, Algeria
| | - Khadidja Habchi
- Nephrology Department, Beni-Messous Teaching Hospital, Faculty of Medicine, University of Algiers, Algiers 16000, Algeria
| | - Messaoud Saidani
- Nephrology Department, Beni-Messous Teaching Hospital, Faculty of Medicine, University of Algiers, Algiers 16000, Algeria
| | - Ines Allam
- Immunology Department, Beni-Messous Teaching Hospital, Faculty of Pharmacy, University of Algiers, Algiers 16000, Algeria
| | - Reda Djidjik
- Immunology Department, Beni-Messous Teaching Hospital, Faculty of Pharmacy, University of Algiers, Algiers 16000, Algeria
| |
Collapse
|
48
|
Johnson SA, Phillips E, Adele S, Longet S, Malone T, Mason C, Stafford L, Jamsen A, Gardiner S, Deeks A, Neo J, Blurton EJ, White J, Ali M, Kronsteiner B, Wilson JD, Skelly DT, Jeffery K, Conlon CP, Goulder P, Consortium PITCH, Carroll M, Barnes E, Klenerman P, Dunachie SJ. Evaluation of QuantiFERON SARS-CoV-2 interferon-γ release assay following SARS-CoV-2 infection and vaccination. Clin Exp Immunol 2023; 212:249-261. [PMID: 36807499 PMCID: PMC10243914 DOI: 10.1093/cei/uxad027] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/18/2023] [Accepted: 02/20/2023] [Indexed: 02/23/2023] Open
Abstract
T cells are important in preventing severe disease from SARS-CoV-2, but scalable and field-adaptable alternatives to expert T-cell assays are needed. The interferon-gamma release assay QuantiFERON platform was developed to detect T-cell responses to SARS-CoV-2 from whole blood with relatively basic equipment and flexibility of processing timelines. Forty-eight participants with different infection and vaccination backgrounds were recruited. Whole blood samples were analysed using the QuantiFERON SARS-CoV-2 assay in parallel with the well-established 'Protective Immunity from T Cells in Healthcare workers' (PITCH) ELISpot, which can evaluate spike-specific T-cell responses. The primary aims of this cross-sectional observational cohort study were to establish if the QuantiFERON SARS-Co-V-2 assay could discern differences between specified groups and to assess the sensitivity of the assay compared with the PITCH ELISpot. The QuantiFERON SARS-CoV-2 distinguished acutely infected individuals (12-21 days post positive PCR) from naïve individuals (P < 0.0001) with 100% sensitivity and specificity for SARS-CoV-2 T cells, whilst the PITCH ELISpot had reduced sensitivity (62.5%) for the acute infection group. Sensitivity with QuantiFERON for previous infection was 12.5% (172-444 days post positive test) and was inferior to the PITCH ELISpot (75%). Although the QuantiFERON assay could discern differences between unvaccinated and vaccinated individuals (55-166 days since second vaccination), the latter also had reduced sensitivity (44.4%) compared to the PITCH ELISpot (66.6%). The QuantiFERON SARS-CoV-2 assay showed potential as a T- cell evaluation tool soon after SARS-CoV-2 infection but has lower sensitivity for use in reliable evaluation of vaccination or more distant infection.
Collapse
Affiliation(s)
- Síle A Johnson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- University of Oxford Medical School, University of Oxford, Oxford, UK
- University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
| | - Eloise Phillips
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Sandra Adele
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Stephanie Longet
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tom Malone
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Chris Mason
- University of Oxford Medical School, University of Oxford, Oxford, UK
| | - Lizzie Stafford
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Department of Experimental Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Anni Jamsen
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Department of Experimental Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Siobhan Gardiner
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Department of Experimental Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Alexandra Deeks
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Department of Experimental Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Janice Neo
- University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
| | - Emily J Blurton
- University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
| | - Jemima White
- University of Oxford Medical School, University of Oxford, Oxford, UK
| | - Muhammed Ali
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Joseph D Wilson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- King’s College Hospital NHS Foundation Trust, London, UK
| | - Dónal T Skelly
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Katie Jeffery
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Christopher P Conlon
- Oxford Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Philip Goulder
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, UK
| | - PITCH Consortium
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Miles Carroll
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Susanna J Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
49
|
Duette G, Lee E, Martins Costa Gomes G, Tungatt K, Doyle C, Stylianou VV, Lee A, Maddocks S, Taylor J, Khanna R, Bull RA, Martinello M, Sandgren KJ, Cunningham AL, Palmer S. Highly Networked SARS-CoV-2 Peptides Elicit T Cell Responses with Enhanced Specificity. Immunohorizons 2023; 7:508-527. [PMID: 37358499 PMCID: PMC10580120 DOI: 10.4049/immunohorizons.2300034] [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: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 06/27/2023] Open
Abstract
Identifying SARS-CoV-2-specific T cell epitope-derived peptides is critical for the development of effective vaccines and measuring the duration of specific SARS-CoV-2 cellular immunity. In this regard, we previously identified T cell epitope-derived peptides within topologically and structurally essential regions of SARS-CoV-2 spike and nucleocapsid proteins by applying an immunoinformatics pipeline. In this study, we selected 30 spike- and nucleocapsid-derived peptides and assessed whether these peptides induce T cell responses and avoid major mutations found in SARS-CoV-2 variants of concern. Our peptide pool was highly specific, with only a single peptide driving cross-reactivity in people unexposed to SARS-COV-2, and immunogenic, inducing a polyfunctional response in CD4+ and CD8+ T cells from COVID-19 recovered individuals. All peptides were immunogenic and individuals recognized broad and diverse peptide repertoires. Moreover, our peptides avoided most mutations/deletions associated with all four SARS-CoV-2 variants of concern while retaining their physicochemical properties even when genetic changes are introduced. This study contributes to an evolving definition of individual CD4+ and CD8+ T cell epitopes that can be used for specific diagnostic tools for SARS-CoV-2 T cell responses and is relevant to the development of variant-resistant and durable T cell-stimulating vaccines.
Collapse
Affiliation(s)
- Gabriel Duette
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Eunok Lee
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Katie Tungatt
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Chloe Doyle
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Vicki V. Stylianou
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ashley Lee
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Susan Maddocks
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Janette Taylor
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Rajiv Khanna
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Rowena A. Bull
- School of Medical Sciences, Faculty of Medicine, UNSW Australia, Sydney, New South Wales, Australia
- The Kirby Institute, UNSW Australia, Sydney, New South Wales, Australia
| | - Marianne Martinello
- The Kirby Institute, UNSW Australia, Sydney, New South Wales, Australia
- Westmead Hospital, Sydney, New South Wales, Australia
- Blacktown & Mount Druitt Hospital, Blacktown, New South Wales, Australia
| | - Kerrie J. Sandgren
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Sarah Palmer
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
50
|
Mohandas S, Jagannathan P, Henrich TJ, Sherif ZA, Bime C, Quinlan E, Portman MA, Gennaro M, Rehman J. Immune mechanisms underlying COVID-19 pathology and post-acute sequelae of SARS-CoV-2 infection (PASC). eLife 2023; 12:e86014. [PMID: 37233729 PMCID: PMC10219649 DOI: 10.7554/elife.86014] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
With a global tally of more than 500 million cases of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections to date, there are growing concerns about the post-acute sequelae of SARS-CoV-2 infection (PASC), also known as long COVID. Recent studies suggest that exaggerated immune responses are key determinants of the severity and outcomes of the initial SARS-CoV-2 infection as well as subsequent PASC. The complexity of the innate and adaptive immune responses in the acute and post-acute period requires in-depth mechanistic analyses to identify specific molecular signals as well as specific immune cell populations which promote PASC pathogenesis. In this review, we examine the current literature on mechanisms of immune dysregulation in severe COVID-19 and the limited emerging data on the immunopathology of PASC. While the acute and post-acute phases may share some parallel mechanisms of immunopathology, it is likely that PASC immunopathology is quite distinct and heterogeneous, thus requiring large-scale longitudinal analyses in patients with and without PASC after an acute SARS-CoV-2 infection. By outlining the knowledge gaps in the immunopathology of PASC, we hope to provide avenues for novel research directions that will ultimately lead to precision therapies which restore healthy immune function in PASC patients.
Collapse
Affiliation(s)
- Sindhu Mohandas
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Prasanna Jagannathan
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford UniversityStanfordUnited States
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California, San FranciscoSan FranciscoUnited States
| | - Zaki A Sherif
- Department of Biochemistry & Molecular Biology, Howard University College of MedicineWashingtonUnited States
| | - Christian Bime
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, University of Arizona College of MedicineTucsonUnited States
| | - Erin Quinlan
- National Center for Complementary and Integrative Health, National Institutes of HealthBethesdaUnited States
| | - Michael A Portman
- Seattle Children’s Hospital, Division of Pediatric Cardiology, Department of Pediatrics, University of WashingtonSeattleUnited States
| | - Marila Gennaro
- Public Health Research Institute and Department of Medicine, Rutgers New Jersey Medical SchoolNewarkUnited States
| | - Jalees Rehman
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of MedicineChicagoUnited States
| |
Collapse
|