51
|
Lu X, Yamasaki S. Current understanding of T cell immunity against SARS-CoV-2. Inflamm Regen 2022; 42:51. [PMID: 36447270 PMCID: PMC9706904 DOI: 10.1186/s41232-022-00242-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022] Open
Abstract
As an important part of adaptive immunity, T cells are indispensable in the defense against pathogens including viruses. SARS-CoV-2 is a new human coronavirus that occurred at the end of 2019 and has caused the COVID-19 pandemic. Nevertheless, most of the infected patients recovered without any antiviral therapies, suggesting an effective immunity developed in the bodies. T cell immunity responds upon SARS-CoV-2 infection or vaccination and plays crucial roles in eliminating the viruses and generating T cell memory. Specifically, a subpopulation of CD4+ T cells could support the production of anti-SARS-CoV-2 antibodies, and cytotoxic CD8+ T cells are also protective against the infection. SARS-CoV-2-recognizing T cells could be detected in SARS-CoV-2-unexposed donors, but the role of these cross-reactive T cells is still in debate. T cell responses could be diverse across individuals, mainly due to the polymorphism of HLAs. Thus, compared to antibodies, T cell responses are generally less affected by the mutations of SARS-CoV-2 variants. Up to now, a huge number of studies on SARS-CoV-2-responsive T cells have been published. In this review, we introduced some major findings addressing the questions in the main aspects about T cell responses elicited by SARS-CoV-2, to summarize the current understanding of COVID-19.
Collapse
Affiliation(s)
- Xiuyuan Lu
- grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Epitope Analysis Team, Center for Advanced Modalities and DDS, Osaka University, Suita, 565-0871 Japan
| | - Sho Yamasaki
- grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Epitope Analysis Team, Center for Advanced Modalities and DDS, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, 565-0871 Japan ,grid.177174.30000 0001 2242 4849Division of Molecular Design, Research Center for Systems Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| |
Collapse
|
52
|
MARSHALL NE, BLANTON MB, DORATT BM, MALHERBE DC, RINCON M, TRUE H, MCDONALD T, BEAUREGARD C, ADATORWOVOR R, MESSAOUDI I. SARS-CoV-2 Vaccine Booster Elicits Robust Prolonged Maternal Antibody Responses and Passive Transfer Via The Placenta And Breastmilk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.11.29.518385. [PMID: 36482972 PMCID: PMC9727762 DOI: 10.1101/2022.11.29.518385] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Background Infection during pregnancy can result in adverse outcomes for both pregnant persons and offspring. Maternal vaccination is an effective mechanism to protect both mother and neonate into post-partum. However, our understanding of passive transfer of antibodies elicited by maternal SARS-CoV-2 mRNA vaccination during pregnancy remains incomplete. Objective We aimed to evaluate the antibody responses engendered by maternal SARS-CoV-2 vaccination following initial and booster doses in maternal circulation and breastmilk to better understand passive immunization of the newborn. Study Design We collected longitudinal blood samples from 121 pregnant women who received SARS-CoV-2 mRNA vaccines spanning from early gestation to delivery followed by collection of blood samples and breastmilk between delivery and 12 months post-partum. During the study, 70% of the participants also received a booster post-partum. Paired maternal plasma, breastmilk, umbilical cord plasma, and newborn plasma samples were tested via enzyme-linked immunosorbent assays (ELISA) to evaluate SARS-CoV-2 specific IgG antibody levels. Results Vaccine-elicited maternal antibodies were detected in both cord blood and newborn blood, albeit at lower levels than maternal circulation, demonstrating transplacental passive immunization. Booster vaccination significantly increased spike specific IgG antibody titers in maternal plasma and breastmilk. Finally, SARS-CoV-2 specific IgG antibodies in newborn blood correlated negatively with days post initial maternal vaccine dose. Conclusion Vaccine-induced maternal SARS-CoV-2 antibodies were passively transferred to the offspring in utero via the placenta and after birth via breastfeeding. Maternal booster vaccination, regardless of gestational age at maternal vaccination, significantly increased antibody levels in breastmilk and maternal plasma, indicating the importance of this additional dose to maximize passive protection against SARS-CoV-2 infection for neonates and infants until vaccination eligibility.
Collapse
Affiliation(s)
- Nicole E. MARSHALL
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR,Corresponding authors: Nicole Marshall and Ilhem Messaoudi, Addresses: Nicole Marshall, Department of Obstetrics and Gynecology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, , Ilhem Messaoudi, Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, 760 Press Ave, Lexington, KY 40536,
| | - Madison B. BLANTON
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY,Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY
| | - Brianna M. DORATT
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY
| | - Delphine C. MALHERBE
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY
| | - Monica RINCON
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR
| | - Heather TRUE
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY,Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY
| | - Taylor MCDONALD
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY
| | - Caroline BEAUREGARD
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY
| | | | - Ilhem MESSAOUDI
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY,Corresponding authors: Nicole Marshall and Ilhem Messaoudi, Addresses: Nicole Marshall, Department of Obstetrics and Gynecology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, , Ilhem Messaoudi, Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, 760 Press Ave, Lexington, KY 40536,
| |
Collapse
|
53
|
Yamaguchi Y, Kato Y, Edahiro R, Søndergaard JN, Murakami T, Amiya S, Nameki S, Yoshimine Y, Morita T, Takeshima Y, Sakakibara S, Naito Y, Motooka D, Liu YC, Shirai Y, Okita Y, Fujimoto J, Hirata H, Takeda Y, Wing JB, Okuzaki D, Okada Y, Kumanogoh A. Consecutive BNT162b2 mRNA vaccination induces short-term epigenetic memory in innate immune cells. JCI Insight 2022; 7:e163347. [PMID: 36282593 PMCID: PMC9746816 DOI: 10.1172/jci.insight.163347] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/07/2022] [Indexed: 12/15/2022] Open
Abstract
Consecutive mRNA vaccinations against SARS-CoV-2 reinforced both innate and adaptive immune responses. However, it remains unclear whether the enhanced innate immune responses are mediated by epigenetic regulation and, if so, whether these effects persist. Using mass cytometry, RNA-Seq, and ATAC-Seq, we show that BNT162b2 mRNA vaccination upregulated antiviral and IFN-stimulated gene expression in monocytes with greater effects after the second vaccination than those after the first vaccination. Transcription factor-binding motif analysis also revealed enriched IFN regulatory factors and PU.1 motifs in accessible chromatin regions. Importantly, although consecutive BNT162b2 mRNA vaccinations boosted innate immune responses and caused epigenetic changes in isolated monocytes, we show that these effects occurred only transiently and disappeared 4 weeks after the second vaccination. Furthermore, single-cell RNA-Seq analysis revealed that a similar gene signature was impaired in the monocytes of unvaccinated patients with COVID-19 with acute respiratory distress syndrome. These results reinforce the importance of the innate immune response in the determination of COVID-19 severity but indicate that, unlike adaptive immunity, innate immunity is not unexpectedly sustained even after consecutive vaccination. This study, which focuses on innate immune memory, may provide novel insights into the vaccine development against infectious diseases.
Collapse
Affiliation(s)
- Yuta Yamaguchi
- Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology and
| | - Yasuhiro Kato
- Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology and
| | - Ryuya Edahiro
- Department of Respiratory Medicine and Clinical Immunology and
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | | | - Teruaki Murakami
- Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology and
| | - Saori Amiya
- Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology and
| | - Shinichiro Nameki
- Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology and
| | - Yuko Yoshimine
- Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology and
| | - Takayoshi Morita
- Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology and
| | - Yusuke Takeshima
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center
| | - Shuhei Sakakibara
- Laboratory of Immune Regulation, WPI Immunology Frontier Research Center
| | - Yoko Naito
- Genome Information Research Center, Research Institute for Microbial Diseases
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases
- Single Cell Genomics, Human Immunology, WPI Immunology Frontier Research Center
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives
| | - Yu-Chen Liu
- Single Cell Genomics, Human Immunology, WPI Immunology Frontier Research Center
| | - Yuya Shirai
- Department of Respiratory Medicine and Clinical Immunology and
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasutaka Okita
- Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology and
| | - Jun Fujimoto
- Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology and
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology and
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology and
| | - James B. Wing
- Human Immunology Team, Center for Infectious Diseases Education and Research
- Single Cell Immunology, Human Immunology, WPI Immunology Frontier Research Center
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases
- Single Cell Genomics, Human Immunology, WPI Immunology Frontier Research Center
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives
- Center for Infectious Diseases Education and Research, and
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives
- Center for Infectious Diseases Education and Research, and
- Statistical Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Atsushi Kumanogoh
- Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology and
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives
- Center for Infectious Diseases Education and Research, and
- Japan Agency for Medical Research and Development – Core Research for Evolutional Science and Technology, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Suita, Japan
| |
Collapse
|
54
|
Yoon BK, Oh TG, Bu S, Seo KJ, Kwon SH, Lee JY, Kim Y, Kim JW, Ahn HS, Fang S. The Peripheral Immune Landscape in a Patient with Myocarditis after the Administration of BNT162b2 mRNA Vaccine. Mol Cells 2022; 45:738-748. [PMID: 35904026 PMCID: PMC9589374 DOI: 10.14348/molcells.2022.0031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/27/2022] [Accepted: 06/09/2022] [Indexed: 12/02/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has posed a serious threat to global public health. A novel vaccine made from messenger RNA (mRNA) has been developed and approved for use at an unprecedented pace. However, an increased risk of myocarditis has been reported after BNT162b2 mRNA vaccination due to unknown causes. In this study, we used single-cell RNA sequencing and single-cell T cell receptor sequencing analyses of peripheral blood mononuclear cells (PBMCs) to describe, for the first time, changes in the peripheral immune landscape of a patient who underwent myocarditis after BNT162b2 vaccination. The greatest changes were observed in the transcriptomic profile of monocytes in terms of the number of differentially expressed genes. When compared to the transcriptome of PBMCs from vaccinated individuals without complications, increased expression levels of IL7R were detected in multiple cell clusters. Overall, results from this study can help advance research into the pathogenesis of BNT162b2-induced myocarditis.
Collapse
Affiliation(s)
- Bo Kyung Yoon
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul 03722, Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Tae Gyu Oh
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Seonghyeon Bu
- Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Uijeongbu St. Mary’s Hospital, Seoul 06591, Korea
- Catholic Research Institute for Intractable Cardiovascular Disease (CRID), College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Kyung Jin Seo
- Department of Pathology, The Catholic University of Korea, Uijeongbu St. Mary’s Hospital, Seoul 06591, Korea
| | - Se Hwan Kwon
- Department of Radiology, Kyung Hee University Medical Center, Seoul 02447, Korea
| | | | | | - Jae-woo Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul 03722, Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Hyo-Suk Ahn
- Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Uijeongbu St. Mary’s Hospital, Seoul 06591, Korea
- Catholic Research Institute for Intractable Cardiovascular Disease (CRID), College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Sungsoon Fang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
- KYNOGEN Co., Suwon 16229, Korea
- Severance Biomedical Science Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
55
|
Dentone C, Fenoglio D, Ponzano M, Cerchiaro M, Altosole T, Franciotta D, Portunato F, Mikulska M, Taramasso L, Magnasco L, Uras C, Magne F, Ferrera F, Scavone G, Signori A, Vena A, Visconti V, Filaci G, Sette A, Grifoni A, Di Biagio A, Bassetti M. Healthcare Worker Study Cohort to Determine the Level and Durability of Cellular and Humoral Immune Responses after Two Doses of SARS-CoV-2 Vaccination. Vaccines (Basel) 2022; 10:1784. [PMID: 36366293 PMCID: PMC9697204 DOI: 10.3390/vaccines10111784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 11/29/2022] Open
Abstract
We prospectively studied immunological response against SARS-CoV-2 after vaccination among healthcare workers without (group A) and with previous infection (group B). The analyses were collected at T0 (before the BNT162b2), T1 (before the second dose), T2 and T6 (1 and 6 months after the second dose). For cellular immune response, the activation-induced cell marker assay was performed with CD4 and CD8 Spike peptide megapools expressed as Stimulation Index. For humoral immune response, we determined antibodies to Spike-1 and nucleocapsid protein. The linear mixed model compared specific times to T0. The CD4+ Spike response overall rate of change was significant at T1 (p = 0.038) and at T2 (p < 0.001), while decreasing at T6. For CD8+ Spike reactivity, the interaction between the time and group was significant (p = 0.0265), and the p value for group comparison was significant at the baseline (p = 0.0030) with higher SI in previously infected subjects. Overall, the anti-S Abs significantly increased from T1 to T6 compared to T0. The group B at T6 retained high anti-S titer (p < 0.001). At T6, in both groups we found a persistent humoral response and a high CD4+ T cell response able to cross recognize SARS-COV-2 variants including epsilon, even if not a circulating virus at that time.
Collapse
Affiliation(s)
- Chiara Dentone
- Infectious Disease Clinic, IRCCS Policlinico San Martino Hospital, Largo Rosanna Benzi, 10, 16132 Genoa, Italy
| | - Daniela Fenoglio
- Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 16126 Genoa, Italy
- Biotherapy Unit, IRCCS Policlinico San Martino, 16132 Genoa, Italy
| | - Marta Ponzano
- Biostatistics Unit, Department of Health Science, University of Genova, 16132 Genova, Italy
| | - Matteo Cerchiaro
- Infectious Diseases Unit, Department of Health Science (DISSAL), University of Genoa, 16126 Genoa, Italy
| | - Tiziana Altosole
- Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 16126 Genoa, Italy
| | - Diego Franciotta
- Autoimmunology Laboratory, IRCCS Policlinico San Martino, 16132 Genoa, Italy
| | - Federica Portunato
- Infectious Disease Clinic, IRCCS Policlinico San Martino Hospital, Largo Rosanna Benzi, 10, 16132 Genoa, Italy
| | - Malgorzata Mikulska
- Infectious Disease Clinic, IRCCS Policlinico San Martino Hospital, Largo Rosanna Benzi, 10, 16132 Genoa, Italy
- Infectious Diseases Unit, Department of Health Science (DISSAL), University of Genoa, 16126 Genoa, Italy
| | - Lucia Taramasso
- Infectious Disease Clinic, IRCCS Policlinico San Martino Hospital, Largo Rosanna Benzi, 10, 16132 Genoa, Italy
| | - Laura Magnasco
- Infectious Disease Clinic, IRCCS Policlinico San Martino Hospital, Largo Rosanna Benzi, 10, 16132 Genoa, Italy
| | - Chiara Uras
- Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 16126 Genoa, Italy
| | - Federica Magne
- Infectious Disease Clinic, IRCCS Policlinico San Martino Hospital, Largo Rosanna Benzi, 10, 16132 Genoa, Italy
| | - Francesca Ferrera
- Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 16126 Genoa, Italy
| | - Graziana Scavone
- Biotherapy Unit, IRCCS Policlinico San Martino, 16132 Genoa, Italy
| | - Alessio Signori
- Biostatistics Unit, Department of Health Science, University of Genova, 16132 Genova, Italy
| | - Antonio Vena
- Infectious Disease Clinic, IRCCS Policlinico San Martino Hospital, Largo Rosanna Benzi, 10, 16132 Genoa, Italy
| | - Valeria Visconti
- Laboratory Unit, IRCCS Policlinico San Martino, 16132 Genoa, Italy
| | - Gilberto Filaci
- Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 16126 Genoa, Italy
- Biotherapy Unit, IRCCS Policlinico San Martino, 16132 Genoa, Italy
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Antonio Di Biagio
- Infectious Disease Clinic, IRCCS Policlinico San Martino Hospital, Largo Rosanna Benzi, 10, 16132 Genoa, Italy
- Infectious Diseases Unit, Department of Health Science (DISSAL), University of Genoa, 16126 Genoa, Italy
| | - Matteo Bassetti
- Infectious Disease Clinic, IRCCS Policlinico San Martino Hospital, Largo Rosanna Benzi, 10, 16132 Genoa, Italy
- Infectious Diseases Unit, Department of Health Science (DISSAL), University of Genoa, 16126 Genoa, Italy
| |
Collapse
|
56
|
Lee HK, Knabl L, Walter M, Furth PA, Hennighausen L. Limited cross-variant immune response from SARS-CoV-2 Omicron BA.2 in naïve but not previously infected outpatients. iScience 2022; 25:105369. [PMID: 36267551 PMCID: PMC9561373 DOI: 10.1016/j.isci.2022.105369] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/12/2022] [Accepted: 10/11/2022] [Indexed: 11/29/2022] Open
Abstract
Omicron is currently the dominant SARS-CoV-2 variant and several sublineages have emerged. Questions remain about the impact of previous SARS-CoV-2 exposure on cross-variant immune responses elicited by the SARS-CoV-2 Omicron sublineage BA.2 compared to BA.1. Here we show that without previous history of COVID-19, BA.2 infection induces a reduced immune response against all variants of concern (VOC) compared to BA.1 infection. The absence of ACE2 binding in sera of previously naïve BA.1 and BA.2 patients indicates a lack of meaningful neutralization. In contrast, anti-spike antibody levels and neutralizing activity greatly increased in the BA.1 and BA.2 patients with a previous history of COVID-19. Transcriptome analyses of peripheral immune cells showed significant differences in immune response and specific antibody generation between BA.1 and BA.2 patients as well as significant differences in expression of specific immune genes. In summary, prior infection status significantly impacts the innate and adaptive immune response against VOC following BA.2 infection.
Collapse
Affiliation(s)
- Hye Kyung Lee
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Mary Walter
- Clinical Core, National Institute of Diabetes, Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Priscilla A Furth
- Departments of Oncology & Medicine, Georgetown University, Washington, DC, USA
| | - Lothar Hennighausen
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
57
|
Cao X, Chen X, Zhu Y, Gou X, Yan K, Yang B, Men D, Liu L, Zhang YA, Cao G. Single-cell transcriptome landscape and antigen receptor dynamic during SARS-CoV-2 vaccination. Genes Dis 2022:S2352-3042(22)00241-0. [PMID: 36097543 PMCID: PMC9454148 DOI: 10.1016/j.gendis.2022.08.020] [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: 04/18/2022] [Revised: 08/04/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
Vaccination by inactivated vaccine is an effective strategy to prevent the COVID-19 pandemic. However, the detailed molecular immune response at single-cell level is poorly understood. In this study, we systematically delineated the landscape of the pre- and post-vaccination single-cell transcriptome, TCR (T cell antigen receptor) and BCR (B cell antigen receptor) expression profile of vaccinated candidates. The bulk TCR sequencing analysis of COVID-19 patients was also performed. Enrichment of a clonal CD8+ T cell cluster expressing specific TCR was identified in both vaccination candidates and COVID-19 patients. These clonal CD8+ T cells showed high expression of cytotoxicity, phagosome and antigen presentation related genes. The cell–cell interaction analysis revealed that monocytes and dendritic cells could interact with these cells and initiate phagocytosis via ICAM1-ITGAM and ITGB2 signaling. Together, our study systematically deciphered the detailed immunological response during SARS-CoV-2 vaccination and infection. It may facilitate understanding the immune response and the T-cell therapy against COVID-19.
Collapse
Affiliation(s)
- Xiaojian Cao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430072, PR China
| | - Xiaohua Chen
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 51015, PR China
| | - Yaqi Zhu
- Department of Laboratory Medicine, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, PR China
| | - Xiaojuan Gou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430072, PR China
| | - Keyi Yan
- Spatial FISH Co. Ltd., Jiangmen 529199, PR China
| | - Bing Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430072, PR China
| | - Dong Men
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, PR China
| | - Lei Liu
- Department of Transfusion Medicine,General Hospital of Central Theater Command, PLA, Wuhan 430070, PR China
| | - Yong-An Zhang
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan 430072, PR China
| | - Gang Cao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430072, PR China.,Bio-Medical College, Huazhong Agricultural University, Wuhan 430072, PR China
| |
Collapse
|
58
|
Qi F, Cao Y, Zhang S, Zhang Z. Single-cell analysis of the adaptive immune response to SARS-CoV-2 infection and vaccination. Front Immunol 2022; 13:964976. [PMID: 36119105 PMCID: PMC9478577 DOI: 10.3389/fimmu.2022.964976] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/10/2022] [Indexed: 12/04/2022] Open
Abstract
Amid the ongoing Coronavirus Disease 2019 (COVID-19) pandemic, vaccination and early therapeutic interventions are the most effective means to combat and control the severity of the disease. Host immune responses to SARS-CoV-2 and its variants, particularly adaptive immune responses, should be fully understood to develop improved strategies to implement these measures. Single-cell multi-omic technologies, including flow cytometry, single-cell transcriptomics, and single-cell T-cell receptor (TCR) and B-cell receptor (BCR) profiling, offer a better solution to examine the protective or pathological immune responses and molecular mechanisms associated with SARS-CoV-2 infection, thus providing crucial support for the development of vaccines and therapeutics for COVID-19. Recent reviews have revealed the overall immune landscape of natural SARS-CoV-2 infection, and this review will focus on adaptive immune responses (including T cells and B cells) to SARS-CoV-2 revealed by single-cell multi-omics technologies. In addition, we explore how the single-cell analyses disclose the critical components of immune protection and pathogenesis during SARS-CoV-2 infection through the comparison between the adaptive immune responses induced by natural infection and by vaccination.
Collapse
Affiliation(s)
- Furong Qi
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Single-Cell Omics Reasearch and Application, Shenzhen, China
| | - Yingyin Cao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Shuye Zhang
- Clinical Center for BioTherapy and Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Single-Cell Omics Reasearch and Application, Shenzhen, China
- Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, China
| |
Collapse
|
59
|
Saresella M, Piancone F, Marventano I, Hernis A, Trabattoni D, Invernizzi M, La Rosa F, Clerici M. Innate immune responses to three doses of the BNT162b2 mRNA SARS-CoV-2 vaccine. Front Immunol 2022; 13:947320. [PMID: 36072604 PMCID: PMC9443429 DOI: 10.3389/fimmu.2022.947320] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
To explore the effects of SARS-CoV-2-mRNA vaccines on innate immune responses we enrolled 58 individuals who received 3 doses of the BNT162b2 vaccine in a longitudinal study; 45 of these individuals had never been SARS-CoV-2 infected. Results showed that vaccination significantly increased: 1) classical and intermediate inflammatory monocytes, 2) CD56bright, CD56dim, and CD56dim/CD16dim NK cells, and 3) IFN-γ+ ;production as well as perforin and granzyme content by NK cells. Vaccination also reduced expression of the NK inhibitory receptor ILT-2, increasing that of the stimulatory molecule 2DS2. These effects were long-lasting and were boosted by every vaccine dose. Notably, ILT-2 expressing NK cells were reduced even more robustly in COVID-19-recovereed vaccines. BNT162b1 mRNA vaccine is known to induce potent adaptive immune responses; results herein show its ability to modulate innate immune responses as well, offering further support to the indication to proceed with worldwide vaccination efforts to end the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Marina Saresella
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- *Correspondence: Marina Saresella,
| | | | | | - Ambra Hernis
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Daria Trabattoni
- Departments of Biomedical and Clinical Sciences L. Sacco, University of Milan, Milan, Italy
| | | | | | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
60
|
Tang Y, Song T, Gao L, Yin S, Ma M, Tan Y, Wu L, Yang Y, Wang Y, Lin T, Li F. A CRISPR-based ultrasensitive assay detects attomolar concentrations of SARS-CoV-2 antibodies in clinical samples. Nat Commun 2022; 13:4667. [PMID: 35945418 PMCID: PMC9361972 DOI: 10.1038/s41467-022-32371-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 07/27/2022] [Indexed: 12/30/2022] Open
Abstract
CRISPR diagnostics are powerful tools for detecting nucleic acids but are generally not deployable for the detection of clinically important proteins. Here, we report an ultrasensitive CRISPR-based antibody detection (UCAD) assay that translates the detection of anti-SARS-CoV-2 antibodies into CRISPR-based nucleic acid detection in a homogeneous solution and is 10,000 times more sensitive than the classic immunoassays. Clinical validation using serum samples collected from the general population (n = 197), demonstrates that UCAD has 100% sensitivity and 98.5% specificity. With ultrahigh sensitivity, UCAD enables the quantitative analysis of serum anti-SARS-CoV-2 levels in vaccinated kidney transplant recipients who are shown to produce "undetectable" anti-SARS-CoV-2 using standard immunoassay. Because of the high sensitivity and simplicity, we anticipate that, upon further clinical validation against large cohorts of clinical samples, UCAD will find wide applications for clinical uses in both centralized laboratories and point-of-care settings.
Collapse
Affiliation(s)
- Yanan Tang
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Turun Song
- Urology Department, Urology Research Institute, Organ Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lu Gao
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Saifu Yin
- Urology Department, Urology Research Institute, Organ Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ming Ma
- Urology Department, Urology Research Institute, Organ Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yun Tan
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Lijuan Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tao Lin
- Urology Department, Urology Research Institute, Organ Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Feng Li
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan, 610064, China.
- Department of Chemistry, Centre for Biotechnology, Brock University, St. Catharines, Ontario, ON, L2S 3A1, Canada.
| |
Collapse
|
61
|
Chen YJ, Cheng PL, Huang WN, Chen HH, Chen HW, Chen JP, Lin CT, Tang KT, Hung WT, Hsieh TY, Chen YH, Chen YM, Hsiao TH. Single-cell RNA sequencing to decipher the immunogenicity of ChAdOx1 nCoV-19/AZD1222 and mRNA-1273 vaccines in patients with autoimmune rheumatic diseases. Front Immunol 2022; 13:920865. [PMID: 35979368 PMCID: PMC9376226 DOI: 10.3389/fimmu.2022.920865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives To investigate the differences between the vector vaccine ChAdOx1 nCoV-19/AZD1222 (Oxford-AstraZeneca) and mRNA-based vaccine mRNA-1273 (Moderna) in patients with autoimmune rheumatic diseases (AIRD), and to explore the cell-cell interactions between high and low anti-SARS-CoV-2 IgG levels in patients with rheumatic arthritis (RA) using single-cell RNA sequencing (scRNA-seq). Methods From September 16 to December 10, 2021, we consecutively enrolled 445 participants (389 patients with AIRD and 56 healthy controls), of whom 236 were immunized with AZD1222 and 209 with mRNA-1273. The serum IgG antibodies to the SARS-CoV-2 receptor-binding domain was quantified by electrochemiluminescence immunoassay at 4-6 weeks after vaccination. Moreover, peripheral blood mononuclear cells (PBMCs) were isolated from RA patients at 4-6 weeks after vaccination for scRNA-seq and further analyzed by CellChat. ScRNA-seq of PBMCs samples from GSE201534 in the Gene Expression Omnibus (GEO) database were also extracted for analysis. Results The anti-SARS-CoV-2 IgG seropositivity rate was 85.34% for AIRD patients and 98.20% for healthy controls. The anti-SARS-CoV-2 IgG level was higher in patients receiving mRNA-1273 than those receiving AZD1222 (β: 35.25, 95% CI: 14.81-55.68, p=0.001). Prednisolone-equivalent dose >5 mg/day and methotrexate use in AIRD patients, and non-anti-tumor necrosis factor-α biologics and Janus kinase inhibitor use in RA patients were associated with inferior immunogenicity. ScRNA-seq revealed CD16-monocytes were predominant in RA patients with high anti-SARS-CoV2-IgG antibodies, and enriched pathways related to antigen presentation via MHC class II were found. HLA-DRA and CD4 interaction was enhanced in high anti-SARS-CoV2-IgG group. Conclusions mRNA-1273 and AZD1222 vaccines exhibited differential immunogenicity in AIRD patients. Enriched pathways related to antigen presentation via MHC class II in CD16-monocytes might be associated with higher anti-SARS-CoV2-IgG level in RA patients and further study is warranted.
Collapse
Affiliation(s)
- Yen-Ju Chen
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Po-Liang Cheng
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wen-Nan Huang
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Hsin-Hua Chen
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung, Taiwan
- Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Public Health and Community Medicine Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Hong-Wei Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jun-Peng Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ching-Tsai Lin
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Kuo-Tung Tang
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Wei-Ting Hung
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Medical Education, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Tsu-Yi Hsieh
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Medical Education, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Hsing Chen
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ming Chen
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Tzu-Hung Hsiao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Public Health, Fu Jen Catholic University, New Taipei City, Taiwan
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
62
|
Xu K, Fan C, Han Y, Dai L, Gao GF. Immunogenicity, efficacy and safety of COVID-19 vaccines: an update of data published by 31 December 2021. Int Immunol 2022; 34:595-607. [PMID: 35778913 PMCID: PMC9278184 DOI: 10.1093/intimm/dxac031] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 06/30/2022] [Indexed: 02/01/2023] Open
Abstract
The unprecedented coronavirus disease 2019 (COVID-19) pandemic has caused a disaster for public health in the last 2 years, without any sign of an ending. Various vaccines were developed rapidly as soon as the outbreak occurred. Clinical trials demonstrated the reactogenicity, immunogenicity and protection efficacy in humans, and some of the vaccines have been approved for clinical use. However, waves of infections such as the recently circulating Omicron variant still occur. Newly emerging variants, especially the variants of concern, and waning humoral responses pose serious challenges to the control of the COVID-19 pandemic. Previously, we summarized the humoral and cellular immunity, safety profiles and protection efficacy of COVID-19 vaccines with clinical data published by 21 May 2021. In this review, we summarize and update the published clinical data of COVID-19 vaccines and candidates up to 31 December 2021.
Collapse
Affiliation(s)
- Kun Xu
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China,Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Hainan, China
| | - Chunxiang Fan
- National Immunization Programme, Chinese Center for Diseases Control and Prevention, Beijing, China
| | - Yuxuan Han
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Lianpan Dai
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Hainan, China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China,CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | | |
Collapse
|
63
|
Lee HK, Go J, Sung H, Kim SW, Walter M, Knabl L, Furth PA, Hennighausen L, Huh JW. Heterologous ChAdOx1-BNT162b2 vaccination in Korean cohort induces robust immune and antibody responses that includes Omicron. iScience 2022; 25:104473. [PMID: 35637788 PMCID: PMC9132682 DOI: 10.1016/j.isci.2022.104473] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/12/2022] [Accepted: 05/20/2022] [Indexed: 01/06/2023] Open
Abstract
Heterologous ChAdOx1-BNT162b2 vaccination induces a stronger immune response than BNT162b2-BNT162b2. Here, we investigated the molecular transcriptome, germline allelic variants of immunoglobulin loci, and anti-Omicron antibody levels in 46 office and lab workers from the Republic of Korea following ChAdOx1-BNT162b2 vaccination. Anti-spike-specific IgG antibody levels against the ancestral SARS-CoV-2 strain increased from 70 AU/ml to 14,000 AU/ml to 142,000 AU/ml one, three and seven days following the second vaccination. Titers against VOC, including Omicron, were two-fold to three-fold lower, yet higher than those measured following BNT162b2-BNT162b2 vaccination. RNA-seq of peripheral immune cells demonstrated activation of interferon pathways with increased IGHV clonal transcripts encoding neutralizing antibodies. scRNA-seq revealed enriched B cell and CD4+ T cell responses in both ChAdOx1-BNT162b2 and BNT162b2-BNT162b2 recipients, but a stronger clonal expansion of memory B cells with ChAdOx1-BNT162b2. In summary, heterologous ChAdOx1-BNT162b2 provides an innate and adaptive immune response that exceeds homologous BNT162b2 vaccination.
Collapse
Affiliation(s)
- Hye Kyung Lee
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jinyoung Go
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Heungsup Sung
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mary Walter
- Clinical Core, National Institute of Diabetes, Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | | | - Priscilla A Furth
- Departments of Oncology & Medicine, Georgetown University, Washington, DC, USA
| | - Lothar Hennighausen
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jin Won Huh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
64
|
Single-cell views of the Plasmodium life cycle. Trends Parasitol 2022; 38:748-757. [DOI: 10.1016/j.pt.2022.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 02/08/2023]
|
65
|
Abstract
Neuroimmunological diseases and their treatment compromise the immune system, thereby increasing the risk of infections and serious illness. Consequently, vaccinations to protect against infections are an important part of the clinical management of these diseases. However, the wide variety of immunotherapies that are currently used to treat neuroimmunological disease — particularly multiple sclerosis and neuromyelitis optica spectrum disorders — can also impair immunological responses to vaccinations. In this Review, we discuss what is known about the effects of various immunotherapies on immunological responses to vaccines and what these effects mean for the safe and effective use of vaccines in patients with a neuroimmunological disease. The success of vaccination in patients receiving immunotherapy largely depends on the specific mode of action of the immunotherapy. To minimize the risk of infection when using immunotherapy, assessment of immune status and exclusion of underlying chronic infections before initiation of therapy are essential. Selection of the required vaccinations and leaving appropriate time intervals between vaccination and administration of immunotherapy can help to safeguard patients. We also discuss the rapidly evolving knowledge of how immunotherapies affect responses to SARS-CoV-2 vaccines and how these effects should influence the management of patients on these therapies during the COVID-19 pandemic. In this Review, the authors discuss how various immunotherapies for neuroimmunological diseases interact with vaccination responses, including responses to SARS-CoV-2 vaccinations, and the implications for the safe and effective use of vaccines in patients with these diseases. Vaccination against infection is an essential part of the management of neuroimmunological diseases. All indicated vaccinations should be administered before initiation of immunotherapy whenever possible; appropriate intervals between vaccination and treatment vary with treatment and vaccination. Inactivated vaccines are considered safe in neuroimmunological diseases but live vaccines are generally contraindicated during immunotherapy. Vaccination responses during immunotherapy can be diminished or abrogated, depending on the treatment and vaccination; antibody titre testing to monitor responses can be considered where appropriate. Vaccinations must be avoided during relapses or exacerbations of neuroimmunological diseases. Vaccination against SARS-CoV-2 is recommended for patients with neuroimmunological disease but some immunotherapies limit the immune response; therefore, timing should be considered carefully.
Collapse
|