1
|
Gao L, Zhang Y, Hu Z, Chen S, Wang Q, Zeng Y, Yin H, Zhao J, Zhan Y, Gao C, Xin Y, Chen B, van der Veen S, Zhao M, Fang D, Lu Q. Microbiota-Derived Inosine Suppresses Systemic Autoimmunity via Restriction of B Cell Differentiation and Migration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409837. [PMID: 40289872 PMCID: PMC12120789 DOI: 10.1002/advs.202409837] [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] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 02/27/2025] [Indexed: 04/30/2025]
Abstract
The role of gut microbiota dysbiosis in systemic lupus erythematosus (SLE) pathogenesis remains elusive. Here, it is shown that fecal microbiota transplantation (FMT) from healthy mice to lupus mice ameliorates lupus-like symptoms. Microbiota reconstitution effectively reduces systemic class switch recombination (CSR) and elevates immunoglobulin heavy chain (IGH) naïve isotype. Microbiota profiling reveals an enrichment of Lactobacillus johnsonii post-FMT, with a significant correlation to purine metabolites. Importantly, the L. johnsonii-derived inosine, an intermediate metabolite in purine metabolism, effectively alleviates lupus pathogenesis in mice. Inosine inhibits B cell differentiation and reduces renal B cell infiltration to protect mice from lupus. At the molecular level, inosine reprograms B cells through the extracellular signal-regulated kinase (ERK)-hypoxia-inducible factor-1alpha (HIF-1α) signaling pathway. Therefore, this study highlights the discovery of a novel microbial metabolite modulating autoimmunity and suggests its potential for innovative microbiome-based therapeutic approaches.
Collapse
Affiliation(s)
- Lingyu Gao
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Yuhan Zhang
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Zhi Hu
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Shengwen Chen
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Qiaolin Wang
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Yong Zeng
- Hunan Key Laboratory of Medical EpigenomicsThe Second Xiangya HospitalCentral South UniversityChangsha410013China
| | - Huiqi Yin
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Junpeng Zhao
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Yijing Zhan
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Changxing Gao
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Yue Xin
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Bing Chen
- Clinical LaboratoryThe Second Hospital of Anhui Medical UniversityHefei230601China
| | - Stijn van der Veen
- Department of Microbiologyand Department of DermatologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310058China
| | - Ming Zhao
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
- Hunan Key Laboratory of Medical EpigenomicsThe Second Xiangya HospitalCentral South UniversityChangsha410013China
| | - Deyu Fang
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoIL60611USA
| | - Qianjin Lu
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
- Hunan Key Laboratory of Medical EpigenomicsThe Second Xiangya HospitalCentral South UniversityChangsha410013China
| |
Collapse
|
2
|
França RKADO, Barros PHA, Silva JM, Fontinele HGC, Maranhão AQ, Brigido MDM. Naive and Memory B Cell BCR Repertoires in Individuals Immunized with an Inactivated SARS-CoV-2 Vaccine. Vaccines (Basel) 2025; 13:393. [PMID: 40333337 PMCID: PMC12031002 DOI: 10.3390/vaccines13040393] [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: 02/24/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic has spurred a global race for a preventive vaccine, with a few becoming available just one year after describing this novel coronavirus disease. Among these are inactivated virus vaccines like CoronaVac (Sinovac Biotech), which are used in several countries to reduce the pandemic's effects. However, its use was associated with low protection, particularly against novel virus variants that quickly appeared in the following months. Vaccines play a crucial role in activating the immune system to combat infections, with Memory B-cells being a key part of this mechanism, eliciting protective neutralizing antibodies. This work focused on studying B-cell memory repertoire after two consecutive doses of CoronaVac. METHODOLOGY Memory B-cells were isolated from five CoronaVac vaccinated and five pre-pandemic individuals and subsequently stimulated in vitro before high-throughput Illumina sequencing of the Heavy Chain Variable repertoire. RESULTS We observed a shift in the VH repertoire with increased HCDR3 length and enrichment of IGVH 3-23, 3-30, 3-7, 3-72, and 3-74 for IgA BCRs and IGHV 4-39 and 4-59 for IgG BCRs. A high expansion of IgA-specific clonal populations was observed in vaccinated individuals relative to pre-pandemic controls, accompanied by shared IgA variable heavy chain (VH) sequences among memory B cells across different vaccine recipients of IgA clones was also observed in vaccinated individuals compared to pre-pandemic controls, with several IgA VH sharing between memory B cells from different vaccines. Moreover, a high convergence was observed among vaccinees and SARS-CoV-2 neutralizing antibody sequences found in the CoV-abDab database. CONCLUSION These data show the ability of CoronaVac to elicit antibodies with characteristics similar to those previously identified as neutralizing antibodies, supporting its protective efficacy. Furthermore, this analysis of the immunological repertoire in the context of viral infections reinforces the importance of immunization in generating convergent antibodies for the antiviral response.
Collapse
Affiliation(s)
- Renato Kaylan Alves de Oliveira França
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasilia 70910-900, DF, Brazil; (R.K.A.d.O.F.); (P.H.A.B.); (H.G.C.F.); (A.Q.M.)
- Molecular Pathology Post-Graduation Program, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Pedro Henrique Aragão Barros
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasilia 70910-900, DF, Brazil; (R.K.A.d.O.F.); (P.H.A.B.); (H.G.C.F.); (A.Q.M.)
- Molecular Biology Post-Graduation Program, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Jacyelle Medeiros Silva
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasilia 70910-900, DF, Brazil; (R.K.A.d.O.F.); (P.H.A.B.); (H.G.C.F.); (A.Q.M.)
| | - Hitallo Guilherme Costa Fontinele
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasilia 70910-900, DF, Brazil; (R.K.A.d.O.F.); (P.H.A.B.); (H.G.C.F.); (A.Q.M.)
- Molecular Pathology Post-Graduation Program, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Andrea Queiroz Maranhão
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasilia 70910-900, DF, Brazil; (R.K.A.d.O.F.); (P.H.A.B.); (H.G.C.F.); (A.Q.M.)
- Molecular Pathology Post-Graduation Program, University of Brasília, Brasilia 70910-900, DF, Brazil
- Molecular Biology Post-Graduation Program, University of Brasília, Brasilia 70910-900, DF, Brazil
- III-Immunology Investigation Institute, National Institute of Science and Technology (iii-INCT), Brasilia 70067-900, DF, Brazil
| | - Marcelo de Macedo Brigido
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasilia 70910-900, DF, Brazil; (R.K.A.d.O.F.); (P.H.A.B.); (H.G.C.F.); (A.Q.M.)
- Molecular Pathology Post-Graduation Program, University of Brasília, Brasilia 70910-900, DF, Brazil
- Molecular Biology Post-Graduation Program, University of Brasília, Brasilia 70910-900, DF, Brazil
- III-Immunology Investigation Institute, National Institute of Science and Technology (iii-INCT), Brasilia 70067-900, DF, Brazil
| |
Collapse
|
3
|
Huang W, de Vries C, Sharma RK, Wangriatisak K, Chatzidionysiou K, Malmström V, Grönwall C. JAK Inhibitors and B Cell Function: A Comparative Study of Their Impact on Plasma Cell Differentiation, Cytokine Production, and Naïve B Cell Activation. Eur J Immunol 2025; 55:e202451437. [PMID: 40098342 PMCID: PMC11914861 DOI: 10.1002/eji.202451437] [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: 08/05/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/19/2025]
Abstract
B cells play a crucial role in autoimmune diseases, as evidenced by autoantibody responses and the effectiveness of B cell-targeted therapies. Janus kinase inhibitors (JAKi), which target downstream signaling of cytokine receptors, are potent rheumatic disease-modifying drugs. However, besides reducing inflammation, JAKi may impact the adaptive immune system. In this study, we examined the effects of JAKi on B-cell function using in vitro cultures and multiparameter flow cytometry. The results show a JAKi-mediated reduction in plasma cell differentiation, primarily by inhibition of memory B-cell stimulation and proliferation. JAKi exposure resulted in stalling R848, IL-2, and IL-21 stimulated B cells in an intermediate activated state with elevated naïve cells displaying increased expression of CXCR5, CD71, CD22, and CD20. In addition, the data demonstrate a moderate JAKi-mediated reduction of B cell TNF and IL-8 cytokine expression following stimulation. Importantly, the efficacy varied greatly between drugs; tofacitinib and upadacitinib (pan JAKi; JAK1i) exhibited the strongest impact, while baricitinib (JAK1/JAK2i) showed donor-dependent variation, and filgotinib (JAK1i) had no effect. All JAKi, except filgotinib, inhibited IL-2 or IL-21-induced STAT3 phosphorylation. Still, filgotinib demonstrated similar inhibition of phospho-STAT5 as other JAKi following IL-21. These findings underscore the therapeutic impact of JAKi through the modulation of B-cell functions.
Collapse
Affiliation(s)
- Wenqi Huang
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| | - Charlotte de Vries
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| | - Ravi Kumar Sharma
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
- Department of Clinical Immunology and RheumatologyAll India Institute of Medical SciencesBilaspurIndia
| | - Kittikorn Wangriatisak
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| | - Katerina Chatzidionysiou
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| | - Vivianne Malmström
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| | - Caroline Grönwall
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| |
Collapse
|
4
|
Lehmann PV, Karulin AY, Becza N, Yao L, Liu Z, Chepke J, Maul-Pavicic A, Wolf C, Köppert S, Valente AV, Gorbachev AV, Tary-Lehmann M, Kirchenbaum GA. Theoretical and practical considerations for validating antigen-specific B cell ImmunoSpot assays. J Immunol Methods 2025; 537:113817. [PMID: 39864733 DOI: 10.1016/j.jim.2025.113817] [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: 02/15/2024] [Revised: 10/17/2024] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
Owing to their ability to reliably detect even very rare antigen-specific B cells in cellular isolates such as peripheral blood mononuclear cells (PBMC), and doing so robustly in a high throughput-compatible manner, B cell ELISPOT/FluoroSpot (collectively "B cell ImmunoSpot") tests have become increasingly attractive for immune monitoring in regulated settings. Presently, there are no guidelines for the qualification and validation of B cell ImmunoSpot assay results. Here, we propose such guidelines, building on the experience acquired from T cell ImmunoSpot testing in an environment adhering to the requirements of regulatory bodies yet taking the unique features of B cell assays into account. A streamlined protocol is proposed that permits the performance of all tests needed for the formal validation of an antigen-specific B cell ImmunoSpot assay in only three experiments, utilizing 2.2 × 107 PBMC per donor. Subsequently, utilizing only 1-2 × 106 PBMC per sample (obtainable from 1 to 2 mL of blood), a validated multiplexed assay enables accurate quantification of the frequency of antigen-specific memory B cell-derived blasts secreting IgM, IgG, IgA or IgE antibodies. Collectively, such multiplexed B cell ImmunoSpot assays offer immense value for B cell immune monitoring programs due to their ease of implementation, scalability, applicability to essentially any antigenic system, economy of PBMC utilization, and last but not least, the high content information gained.
Collapse
Affiliation(s)
- Paul V Lehmann
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Alexey Y Karulin
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Noémi Becza
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Lingling Yao
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Zhigang Liu
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Jack Chepke
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Andrea Maul-Pavicic
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Carla Wolf
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Sebastian Köppert
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Alexis V Valente
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Anton V Gorbachev
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Magdalena Tary-Lehmann
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Greg A Kirchenbaum
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| |
Collapse
|
5
|
Silva JM, França RKADO, Barros PH, Fontinele HGC, Fonseca SG, Brigido MM, Maranhão AQ. Rescuing pathogen-specific memory B-cell from PBMC of prior Zika virus-infected individuals. Immunol Lett 2025; 271:106944. [PMID: 39542046 DOI: 10.1016/j.imlet.2024.106944] [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: 05/14/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Immunological memory, a fundamental immune system mechanism, is instrumental in long-term protection. Successful vaccines can elicit and sustain immunological memory against pathogens for the long term. Memory B cells (MBC) are key players in secondary responses due to their longevity and rapid differentiation into high-affinity antibody-secreting cells upon second antigen exposure. However, the availability of circulating MBCs is limited. Here we describe a protocol, which presents a straightforward and practical method for activating and expanding Zika virus (ZIKV) specific MBC. PBMCs collected from individuals who had been infected with ZIKV two years prior were cultured by supplementing with IL-2 and R848, a TLR-7/8 agonist, and then pulsed with inactivated virus. After seven days, this stimulation led to a notable rise in virus-specific functional MBC, as evidenced by a significant increase in the production of anti-ZIKV IgG. Importantly, the ZIKV pulse did not induce changes in the PBMC culture of individuals without a history of ZIKV infection. These findings demonstrate that virus-specific MBC can be expanded in vitro, even using PBMC cultures from individuals infected years before. Therefore, our protocol is a practical and effective tool for studies that require a larger number of human MBCs from previously infected individuals that are functional and specific to the pathogen under investigation.
Collapse
Affiliation(s)
- Jacyelle Medeiros Silva
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasília, Brazil; Molecular Biology Post-Graduation Program, UnB, Brazil
| | - Renato Kaylan Alves de Oliveira França
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasília, Brazil; Molecular Pathology Post-Graduation Program, UnB, Brazil
| | - Pedro Henrique Barros
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasília, Brazil; Molecular Biology Post-Graduation Program, UnB, Brazil
| | - Hitallo Guilherme Costa Fontinele
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasília, Brazil; Molecular Pathology Post-Graduation Program, UnB, Brazil
| | - Simone Gonçalves Fonseca
- Institute of Tropical Pathology and Public Health, University of Goiás, Goiás, Brazil; iii- Institute for Investigation in Immunology - INCT-iii - CNPq/MCT, Brazil
| | - Marcelo Macedo Brigido
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasília, Brazil; Molecular Biology Post-Graduation Program, UnB, Brazil; Molecular Pathology Post-Graduation Program, UnB, Brazil; iii- Institute for Investigation in Immunology - INCT-iii - CNPq/MCT, Brazil.
| | - Andrea Queiroz Maranhão
- Department of Cellular Biology, Institute of Biological Science, University of Brasília, Brasília, Brazil; Molecular Biology Post-Graduation Program, UnB, Brazil; Molecular Pathology Post-Graduation Program, UnB, Brazil; iii- Institute for Investigation in Immunology - INCT-iii - CNPq/MCT, Brazil
| |
Collapse
|
6
|
Mavrangelos C, Wijenayaka A, Sales KJ, Hughes PA. Combining magnetically isolated CD45 cells with serum maintains intact drug responsiveness for ELISpot analysis in clinical trials. Immunohorizons 2025; 9:vlae012. [PMID: 39882960 PMCID: PMC11841966 DOI: 10.1093/immhor/vlae012] [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: 09/11/2024] [Accepted: 12/04/2024] [Indexed: 01/31/2025] Open
Abstract
Enzyme-linked immunosorbent spot analysis is frequently used to investigate immune responsiveness during clinical trials. However, ELISpot classically utilizes peripheral blood mononuclear cell isolates from whole blood, requiring relatively high blood draw volumes and removing both granulocytes and bound drug. Here, we describe a novel protocol whereby CD45 cells are magnetically isolated from human whole blood and co-incubated with serum isolated from the same subject. Infliximab is a well characterized anti-tumor necrosis factor α (TNF-α) antibody in clinical use since the late 1990s. We demonstrated that TNF-α inhibition by infliximab in spiked whole blood is lost on peripheral blood mononuclear cell isolation but remains in serum, and that combining serum from infliximab spiked whole blood with magnetically isolated CD45 immune cells inhibited PMA/ionomycin-stimulated TNF-α secretion. This novel protocol has important implications for enzyme-linked immunosorbent spot analysis in clinical trials in which blood volume is limited, and keeping drug responses intact provides critical information.
Collapse
Affiliation(s)
| | - Asiri Wijenayaka
- Agilex Biolabs, Adelaide, South Australia, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Kurt J Sales
- Agilex Biolabs, Adelaide, South Australia, Australia
| | - Patrick A Hughes
- Agilex Biolabs, Adelaide, South Australia, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
7
|
Huang Y, Li L, Chen R, Yu L, Zhao S, Jia Y, Dou Y, Zhang Z, An Y, Tang X, Zhao X, Zhou L. Heterogeneous phenotype of a Chinese Familial WHIM syndrome with CXCR4 V340fs gain-of-function mutation. Front Immunol 2024; 15:1460990. [PMID: 39575248 PMCID: PMC11578956 DOI: 10.3389/fimmu.2024.1460990] [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: 07/07/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Background WHIM syndrome is a rare, autosomal dominant inborn error of immunity characterized by warts, hypogammaglobulinemia, infection, and myelokathexis. It is caused mainly by heterozygous mutations at the C-terminus of the C-X-C chemokine receptor type 4 (CXCR4) gene. Methods We described the detailed clinical, genetic, immunological and treatment characteristic of four WHIM patients from a single Chinese family. Results Here, we report four patients from a family carrying a variant of CXCR4 (c.1016_1017dupCT), which introduces a frameshift at codon V340, resulting in an extension of 14 amino acids (p.V340L fs*27). We provide and in-depth analysis of their clinical, genetic, immunological and treatment characteristic, noting that these patients exhibited an atypical clinical phenotype when compared to reported CXCR4R334X patients. Additionally, the frameshift variant CXCR4V340fs led to impaired receptor downregulation in patients' PBMCs, and in HEK293T cells transfected with the variant plasmids. Conclusions Our study provided detailed clinical features of four CXCR4V340fs WHIM patients from one Chinese family who presented atypical phenotype and enrich the spectrum of WHIM syndrome.
Collapse
Affiliation(s)
- Yu Huang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Hematology Oncology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Lu Li
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Ran Chen
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Lang Yu
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Shunkai Zhao
- Department of Biology, School of Arts and Sciences, Tufts University, Medford, MA, United States
| | - Yanjun Jia
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Dou
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Hematology Oncology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiyong Zhang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Rheumatism and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Yunfei An
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Rheumatism and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xuemei Tang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Rheumatism and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Zhao
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Rheumatism and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Lina Zhou
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Rohrbacher S, Seefried S, Hartmannsberger B, Annabelle R, Appeltshauser L, Arlt FA, Brämer D, Dresel C, Dorst J, Elmas Z, Franke C, Geis C, Högen T, Krause S, Marziniak M, Mäurer M, Prüss H, Schoeberl F, Schrank B, Steen C, Teichtinger H, Thieme A, Wessely L, Zernecke A, Sommer C, Doppler K. Different Patterns of Autoantibody Secretion by Peripheral Blood Mononuclear Cells in Autoimmune Nodopathies. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200295. [PMID: 39173087 PMCID: PMC11379437 DOI: 10.1212/nxi.0000000000200295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
BACKGROUND AND OBJECTIVES Autoimmune nodopathies with antibodies against the paranodal proteins show a distinct phenotype of a severe sensorimotor neuropathy. In some patients, complete remission can be achieved after treatment with rituximab whereas others show a chronic course. For optimal planning of treatment, predicting the course of disease and therapeutic response is crucial. METHODS We stimulated peripheral blood mononuclear cells in vitro to find out whether secretion of specific autoantibodies may be a predictor of the course of disease and response to rituximab. RESULTS Three patterns could be identified: In most patients with anti-Neurofascin-155-, anti-Contactin-1-, and anti-Caspr1-IgG4 autoantibodies, in vitro production of autoantibodies was detected, indicating autoantigen-specific memory B cells and short-lived plasma cells/plasmablasts as the major source of autoantibodies. These patients generally showed a good response to rituximab. In a subgroup of patients with anti-Neurofascin-155-IgG4 autoantibodies and insufficient response to rituximab, no in vitro autoantibody production was found despite high serum titers, indicating autoantibody secretion by long-lived plasma cells outside the peripheral blood. In the patients with anti-pan-Neurofascin autoantibodies-all with a monophasic course of disease-no in vitro autoantibody production could be measured, suggesting a lack of autoantigen-specific memory B cells. In some of them, autoantibody production by unstimulated cells was detectable, presumably corresponding to high amounts of autoantigen-specific plasmablasts-well in line with a severe but monophasic course of disease. DISCUSSION Our data suggest that different B-cell responses may occur in autoimmune nodopathies and may serve as markers of courses of disease and response to rituximab.
Collapse
Affiliation(s)
- Sophia Rohrbacher
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Sabine Seefried
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Beate Hartmannsberger
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Rosa Annabelle
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Luise Appeltshauser
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Friederike A Arlt
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Dirk Brämer
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Christian Dresel
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Johannes Dorst
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Zeynep Elmas
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Christiana Franke
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Christian Geis
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Tobias Högen
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Sabine Krause
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Martin Marziniak
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Mathias Mäurer
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Harald Prüss
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Florian Schoeberl
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Bertold Schrank
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Claudia Steen
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Helena Teichtinger
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Andrea Thieme
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Lena Wessely
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Alma Zernecke
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Claudia Sommer
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Kathrin Doppler
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| |
Collapse
|
9
|
Negi S, Rutman AK, Saw CL, Paraskevas S, Tchervenkov J. Pretransplant, Th17 dominant alloreactivity in highly sensitized kidney transplant candidates. FRONTIERS IN TRANSPLANTATION 2024; 3:1336563. [PMID: 38993777 PMCID: PMC11235243 DOI: 10.3389/frtra.2024.1336563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/21/2024] [Indexed: 07/13/2024]
Abstract
Introduction Sensitization to donor human leukocyte antigen (HLA) molecules prior to transplantation is a significant risk factor for delayed access to transplantation and to long-term outcomes. Memory T cells and their cytokines play a pivotal role in shaping immune responses, thereby increasing the risk of allograft rejection among highly sensitized patients. This study aims to elucidate the precise contribution of different CD4+ memory T cell subsets to alloreactivity in highly sensitized (HS) kidney transplant recipients. Methods and results Stimulation of peripheral blood mononuclear cells (PBMC) with various polyclonal stimulating agents to assess non-specific immune responses revealed that HS patients exhibit elevated immune reactivity even before kidney transplantation, compared to non-sensitized (NS) patients. HS patients' PBMC displayed higher frequencies of CD4+ T cells expressing IFNγ, IL4, IL6, IL17A, and TNFα and secreted relatively higher levels of IL17A and IL21 upon stimulation with PMA/ionomycin. Additionally, PBMC from HS patients stimulated with T cell stimulating agent phytohemagglutinin (PHA) exhibited elevated expression levels of IFNγ, IL4 and, IL21. On the other hand, stimulation with a combination of resiquimod (R848) and IL2 for the activation of memory B cells demonstrated higher expression of IL17A, TNFα and IL21, as determined by quantitative real-time PCR. A mixed leukocyte reaction (MLR) assay, employing third-party donor antigen presenting cells (APCs), was implemented to evaluate the direct alloreactive response. HS patients demonstrated notably higher frequencies of CD4+ T cells expressing IL4, IL6 and IL17A. Interestingly, APCs expressing recall HLA antigens triggered a stronger Th17 response compared to APCs lacking recall HLA antigens in sensitized patients. Furthermore, donor APCs induced higher activation of effector memory T cells in HS patients as compared to NS patients. Conclusion These results provide an assessment of pretransplant alloreactive T cell subsets in highly sensitized patients and emphasize the significance of Th17 cells in alloimmune responses. These findings hold promise for the development of treatment strategies tailored to sensitized kidney transplant recipients, with potential clinical implications.
Collapse
Affiliation(s)
- Sarita Negi
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Human Islet Transplantation Laboratory, McGill University Health Centre, Montréal, QC, Canada
| | | | - Chee Loong Saw
- HLA Laboratory, Division of Hematology, McGill University Health Centre, Montréal, QC, Canada
| | - Steven Paraskevas
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Human Islet Transplantation Laboratory, McGill University Health Centre, Montréal, QC, Canada
- Department of Surgery, McGill University, Montréal, QC, Canada
- Division of General Surgery and Multi-Organ Transplant Program, Department of Surgery, McGill University Health Centre, Montréal, QC, Canada
| | - Jean Tchervenkov
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Surgery, McGill University, Montréal, QC, Canada
- Division of General Surgery and Multi-Organ Transplant Program, Department of Surgery, McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
10
|
Lee C, Imran I, Thomas S, Nouri-Shirazi M. A comprehensive method for the phenotypical and functional characterization of recalled human memory B and T cells specific to vaccine antigens. J Immunol Methods 2024; 527:113650. [PMID: 38428517 DOI: 10.1016/j.jim.2024.113650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/10/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
Current methodologies for assessing vaccine effectiveness and longevity primarily center on measuring vaccine-induced neutralizing antibodies in serum or plasma. However, these methods overlook additional parameters such as the presence of memory B cells, even as antibody levels wane, and the pivotal role played by memory T cells in shaping antigen-specific memory B cell responses. Several studies have employed a combination of polyclonal activators, such as CpG and R848, along with various cytokines to provoke the recall of memory B cells from peripheral blood mononuclear cells (PBMCs) into antibody-secreting cells (ASCs). Other studies have examined the use of live attenuated viruses to stimulate antigen-specific memory T cells within PBMCs into effector T cells that produce Th1/Th2 cytokines. However, these studies have not fully elucidated the distinct effects of these polyclonal activators on individual subsets, nor have they evaluated whether the vaccine antigen alone is sufficient to trigger the recall of memory T cells. Thus, in this study, we directly compared the capacity of two B cell polyclonal activators to induce the transition of existing vaccine-specific memory cells present in peripheral blood samples into ASCs. Simultaneously, we also assessed the transition of existing memory T cells into effector subsets in response to vaccine antigens. Our findings demonstrate that both polyclonal activator combinations, CpG with IL-6 and IL-15, as well as R848 with IL-2, effectively induce the terminal differentiation of memory B cells into ASCs. Notably, CpG treatment preferentially expanded naïve and non-class-switched B cells, while R848 expanded class-switched memory cells, plasmablasts, and plasma cells. Consequently, R848 treatment led to a greater overall production of total and antigen-specific IgG immunoglobulins. Additionally, the exposure of isolated PBMCs to vaccine antigens alone proved sufficient for recalling the rare antigen-specific memory T cells into effector subsets, predominantly consisting of IFN-γ-producing CD4 T cells and TNF-β-producing CD8 T cells. This study not only establishes a rationale for the selection of methods to expand and detect antigen-specific lymphocyte subsets but also presents a means to quantify vaccine effectiveness by correlating serum antibody levels with preexisting memory cells within peripheral blood samples.
Collapse
Affiliation(s)
- Czdari Lee
- Florida Atlantic University, Charles E. Schmidt College of Medicine, Department of Medicine, 777 Glades Road, PO Box 3091, Boca Raton, FL 33431, USA
| | - Imtisal Imran
- Florida Atlantic University, Charles E. Schmidt College of Medicine, Department of Medicine, 777 Glades Road, PO Box 3091, Boca Raton, FL 33431, USA
| | - Sara Thomas
- Florida Atlantic University, Charles E. Schmidt College of Medicine, Department of Medicine, 777 Glades Road, PO Box 3091, Boca Raton, FL 33431, USA
| | - Mahyar Nouri-Shirazi
- Florida Atlantic University, Charles E. Schmidt College of Medicine, Department of Medicine, 777 Glades Road, PO Box 3091, Boca Raton, FL 33431, USA.
| |
Collapse
|
11
|
Huang Z, Zhuang X, Liu L, Zhao J, Ma S, Si X, Zhu Z, Wu F, Jin N, Tian M, Song W, Chen X. Modularized viromimetic polymer nanoparticle vaccines (VPNVaxs) to elicit durable and effective humoral immune responses. Natl Sci Rev 2024; 11:nwad310. [PMID: 38312378 PMCID: PMC10833449 DOI: 10.1093/nsr/nwad310] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/27/2023] [Accepted: 11/23/2023] [Indexed: 02/06/2024] Open
Abstract
Virus-like particle (VLP) vaccines had shown great potential during the COVID-19 pandemic, and was thought to be the next generation of antiviral vaccine technology due to viromimetic structures. However, the time-consuming and complicated processes in establishing a current recombinant-protein-based VLP vaccine has limited its quick launch to the out-bursting pandemic. To simplify and optimize VLP vaccine design, we herein report a kind of viromimetic polymer nanoparticle vaccine (VPNVax), with subunit receptor-binding domain (RBD) proteins conjugated to the surface of polyethylene glycol-b-polylactic acid (PEG-b-PLA) nanoparticles for vaccination against SARS-CoV-2. The preparation of VPNVax based on synthetic polymer particle and chemical post-conjugation makes it possible to rapidly replace the antigens and construct matched vaccines at the emergence of different viruses. Using this modular preparation system, we identified that VPNVax with surface protein coverage of 20%-25% had the best immunostimulatory activity, which could keep high levels of specific antibody titers over 5 months and induce virus neutralizing activity when combined with an aluminum adjuvant. Moreover, the polymer nano-vectors could be armed with more immune-adjuvant functions by loading immunostimulant agents or chemical chirality design. This VPNVax platform provides a novel kind of rapidly producing and efficient vaccine against different variants of SARS-CoV-2 as well as other viral pandemics.
Collapse
Affiliation(s)
- Zichao Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Xinyu Zhuang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Liping Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Jiayu Zhao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Sheng Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Zhenyi Zhu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Fan Wu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Ningyi Jin
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Mingyao Tian
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| |
Collapse
|
12
|
Swanson IM, Haralambieva IH, Rasche MM, Ovsyannikova IG, Kennedy RB. Frequencies of SARS-CoV-2 Spike Protein-Specific Memory B Cells in Human PBMCs, Quantified by ELISPOT Assay. Methods Mol Biol 2024; 2768:153-166. [PMID: 38502393 DOI: 10.1007/978-1-0716-3690-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Vaccination against SARS-CoV-2 with coronavirus vaccines that elicit protective immune responses is critical to the prevention of severe disease and mortality associated with SARS-CoV-2 infection. Understanding the adaptive immune responses to SARS-CoV-2 infection and/or vaccination will continue to aid in the development of next-generation vaccines. Studies have shown the important role of SARS-CoV-2-specific antibodies for both disease resolution and prevention of COVID-19 serious sequelae following vaccination. However, antibody responses are short-lived, highlighting the importance of studying antigen-specific B-cell responses to better understand durable immunity and immunologic memory. Since the spike protein is the main target of antibody-producing B cells, we developed a SARS-CoV-2 memory B cell ELISPOT assay to measure the frequencies of spike-specific B cells after COVID-19 infection and/or vaccination. Here, we describe in detail the methodology for using this ELISPOT assay to quantify SARS-CoV-2 spike-specific memory B cells produced by infection and/or vaccination in human PBMC samples. Application of this assay may help better understand and predict SARS-CoV-2 recall immune responses and to develop potential B cell correlates of protection at the methodological level.
Collapse
Affiliation(s)
- Ilya M Swanson
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
13
|
Amash A, Volkers G, Farber P, Griffin D, Davison KS, Goodman A, Tonikian R, Yamniuk A, Barnhart B, Jacobs T. Developability considerations for bispecific and multispecific antibodies. MAbs 2024; 16:2394229. [PMID: 39189686 DOI: 10.1080/19420862.2024.2394229] [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: 06/13/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 08/28/2024] Open
Abstract
Bispecific antibodies (bsAb) and multispecific antibodies (msAb) encompass a diverse variety of formats that can concurrently bind multiple epitopes, unlocking mechanisms to address previously difficult-to-treat or incurable diseases. Early assessment of candidate developability enables demotion of antibodies with low potential and promotion of the most promising candidates for further development. Protein-based therapies have a stringent set of developability requirements in order to be competitive (e.g. high-concentration formulation, and long half-life) and their assessment requires a robust toolkit of methods, few of which are validated for interrogating bsAbs/msAbs. Important considerations when assessing the developability of bsAbs/msAbs include their molecular format, likelihood for immunogenicity, specificity, stability, and potential for high-volume production. Here, we summarize the critical aspects of developability assessment, and provide guidance on how to develop a comprehensive plan tailored to a given bsAb/msAb.
Collapse
Affiliation(s)
- Alaa Amash
- AbCellera Biologics Inc, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | | - Tim Jacobs
- AbCellera Biologics Inc, Vancouver, BC, Canada
| |
Collapse
|
14
|
Abu-Raya B, Esser MJ, Nakabembe E, Reiné J, Amaral K, Diks AM, Imede E, Way SS, Harandi AM, Gorringe A, Le Doare K, Halperin SA, Berkowska MA, Sadarangani M. Antibody and B-cell Immune Responses Against Bordetella Pertussis Following Infection and Immunization. J Mol Biol 2023; 435:168344. [PMID: 37926426 DOI: 10.1016/j.jmb.2023.168344] [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: 08/30/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Neither immunization nor recovery from natural infection provides life-long protection against Bordetella pertussis. Replacement of a whole-cell pertussis (wP) vaccine with an acellular pertussis (aP) vaccine, mutations in B. pertussis strains, and better diagnostic techniques, contribute to resurgence of number of cases especially in young infants. Development of new immunization strategies relies on a comprehensive understanding of immune system responses to infection and immunization and how triggering these immune components would ensure protective immunity. In this review, we assess how B cells, and their secretory products, antibodies, respond to B. pertussis infection, current and novel vaccines and highlight similarities and differences in these responses. We first focus on antibody-mediated immunity. We discuss antibody (sub)classes, elaborate on antibody avidity, ability to neutralize pertussis toxin, and summarize different effector functions, i.e. ability to activate complement, promote phagocytosis and activate NK cells. We then discuss challenges and opportunities in studying B-cell immunity. We highlight shared and unique aspects of B-cell and plasma cell responses to infection and immunization, and discuss how responses to novel immunization strategies better resemble those triggered by a natural infection (i.e., by triggering responses in mucosa and production of IgA). With this comprehensive review, we aim to shed some new light on the role of B cells and antibodies in the pertussis immunity to guide new vaccine development.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.
| | - Mirjam J Esser
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Eve Nakabembe
- Centre for Neonatal and Paediatric Infectious Diseases Research, St George's, University of London, Cranmer Terrace, London SW17 0RE, UK; Department of Obstetrics and Gynaecology, Makerere University College of Health Sciences, Upper Mulago Hill Road, Kampala, P.O. Box 7072, Uganda
| | - Jesús Reiné
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom; Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Kyle Amaral
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Annieck M Diks
- Department of Immunology, Leiden University Medical Center, Albinusdreef 2, Leiden ZA 2333, the Netherlands
| | - Esther Imede
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Sing Sing Way
- Department of Pediatrics, Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Ali M Harandi
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Andrew Gorringe
- UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| | - Kirsty Le Doare
- Centre for Neonatal and Paediatric Infectious Diseases Research, St George's, University of London, Cranmer Terrace, London SW17 0RE, UK; Makerere University-Johns Hopkins University Research Collaboration, MU-JHU, Upper Mulago Hill, Kampala, P.O. Box 23491, Uganda
| | - Scott A Halperin
- Canadian Center for Vaccinology, Departments of Pediatrics and Microbiology and Immunology, Dalhousie University, Izaak Walton Killam Health Centre, and Nova Scotia Health Authority, Halifax, NS, Canada
| | - Magdalena A Berkowska
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
15
|
Chen Y, Zhou S, Pradhan K, Chernyak N, Kofman E, Zhang F, Kim SY, Seghezzi W, Willingham A, Seganish WM, Bhagwat B, Han JH. Development and application of an in vitro assay to assess target-independent B-cell activation by targeted TLR7 immune agonists. J Immunol Methods 2023; 522:113553. [PMID: 37661047 DOI: 10.1016/j.jim.2023.113553] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
Targeted immune agonist (TIA) comprising a TLR7 agonist conjugated to tumor-targeting antibodies have been shown to induce potent anti-tumor responses in various preclinical models. However, the clinical proof-of-concept of a TIA has been hampered by systemic dose-limiting immune-related toxicities, including rapid induction of anti-drug antibodies in patients. We have developed ELISPOT-based assay to measure activation of antibody-secreting cells (ASCs), intended to simulate the interaction between TIA and peripheral B cells as a tool to pre-clinically de-risk tumor target-independent peripheral B-cell activation by TIA. This method has proven to be robust and has fast turn-around time to evaluate the induction of spontaneous B-cell activation by TIA in a tumor target- and FcγR-independent manner. This novel ASC assay platform may serve as a preclinical tool to de-risk TIAs that can potentially induce immune-related adverse effects in the clinic.
Collapse
|
16
|
Stensland ZC, Magera CA, Broncucia H, Gomez BD, Rios-Guzman NM, Wells KL, Nicholas CA, Rihanek M, Hunter MJ, Toole KP, Gottlieb PA, Smith MJ. Identification of an anergic BND cell-derived activated B cell population (BND2) in young-onset type 1 diabetes patients. J Exp Med 2023; 220:e20221604. [PMID: 37184563 PMCID: PMC10192302 DOI: 10.1084/jem.20221604] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 03/15/2023] [Accepted: 04/21/2023] [Indexed: 05/16/2023] Open
Abstract
Recent evidence suggests a role for B cells in the pathogenesis of young-onset type 1 diabetes (T1D), wherein rapid progression occurs. However, little is known regarding the specificity, phenotype, and function of B cells in young-onset T1D. We performed a cross-sectional analysis comparing insulin-reactive to tetanus-reactive B cells in the blood of T1D and controls using mass cytometry. Unsupervised clustering revealed the existence of a highly activated B cell subset we term BND2 that falls within the previously defined anergic BND subset. We found a specific increase in the frequency of insulin-reactive BND2 cells in the blood of young-onset T1D donors, which was further enriched in the pancreatic lymph nodes of T1D donors. The frequency of insulin-binding BND2 cells correlated with anti-insulin autoantibody levels. We demonstrate BND2 cells are pre-plasma cells and can likely act as APCs to T cells. These findings identify an antigen-specific B cell subset that may play a role in the rapid progression of young-onset T1D.
Collapse
Affiliation(s)
- Zachary C. Stensland
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Christopher A. Magera
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hali Broncucia
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Brittany D. Gomez
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nasha M. Rios-Guzman
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kristen L. Wells
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Catherine A. Nicholas
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marynette Rihanek
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Maya J. Hunter
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kevin P. Toole
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Peter A. Gottlieb
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Mia J. Smith
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
17
|
Conarty JP, Wieland A. The Tumor-Specific Immune Landscape in HPV+ Head and Neck Cancer. Viruses 2023; 15:1296. [PMID: 37376596 DOI: 10.3390/v15061296] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Human papillomaviruses (HPVs) are the causative agent of several anogenital cancers as well as head and neck cancers, with HPV+ head and neck squamous cell carcinoma (HNSCC) becoming a rapidly growing public health issue in the Western world. Due its viral etiology and potentially its subanatomical location, HPV+ HNSCC exhibits an immune microenvironment which is more inflamed and thus distinct from HPV-negative HNSCC. Notably, the antigenic landscape in most HPV+ HNSCC tumors extends beyond the classical HPV oncoproteins E6/7 and is extensively targeted by both the humoral and cellular arms of the adaptive immune system. Here, we provide a comprehensive overview of HPV-specific immune responses in patients with HPV+ HNSCC. We highlight the localization, antigen specificity, and differentiation states of humoral and cellular immune responses, and discuss their similarities and differences. Finally, we review currently pursued immunotherapeutic treatment modalities that attempt to harness HPV-specific immune responses for improving clinical outcomes in patients with HPV+ HNSCC.
Collapse
Affiliation(s)
- Jacob P Conarty
- Department of Otolaryngology, The Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Andreas Wieland
- Department of Otolaryngology, The Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
18
|
Tang Z, Yu P, Guo Q, Chen M, Lei Y, Zhou L, Mai W, Chen L, Deng M, Kong W, Niu C, Xiong X, Li W, Chen C, Lai C, Wang Q, Li B, Ji T. Clinical characteristics and host immunity responses of SARS-CoV-2 Omicron variant BA.2 with deletion of ORF7a, ORF7b and ORF8. Virol J 2023; 20:106. [PMID: 37248496 DOI: 10.1186/s12985-023-02066-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/08/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND The pathogenicity and virulence of the Omicron strain have weakened significantly pathogenesis of Omicron variants. Accumulating data indicated accessory proteins play crucial roles in host immune evasion and virus pathogenesis of SARS-CoV-2. Therefore, the impact of simultaneous deletion of accessory protein ORF7a, ORF7b and ORF8 on the clinical characteristics and specific immunity in Omicron breakthrough infected patients (BIPs) need to be verified. METHODS Herein, plasma cytokines were identified using a commercial Multi-cytokine detection kit. Enzyme-linked immunosorbent assay and pseudovirus neutralization assays were utilized to determine the titers of SARS-CoV-2 specific binding antibodies and neutralizing antibodies, respectively. In addition, an enzyme-linked immunospot assay was used to quantify SARS-CoV-2 specific T cells and memory B cells. RESULTS A local COVID-19 outbreak was caused by the Omicron BA.2 variant, which featured a deletion of 871 base pairs (∆871 BA.2), resulting in the removal of ORF7a, ORF7b, and ORF8. We found that hospitalized patients with ∆871 BA.2 had significantly shorter hospital stays than those with wild-type (WT) BA.2. Plasma cytokine levels in both ∆871 BA.2 and WT BA.2 patients were within the normal range of reference, and there was no notable difference in the titers of SARS-CoV-2 ancestor or Omicron-specific binding IgG antibodies, neutralizing antibody titers, effector T cells, and memory B cells frequencies between ∆871 BA.2 and WT BA.2 infected adult patients. However, antibody titers in ∆871 BA.2 infected adolescents were higher than in adults. CONCLUSIONS The simultaneous deletion of ORF7a, ORF7b, and ORF8 facilitates the rapid clearance of the BA.2 variant, without impacting cytokine levels or affecting SARS-CoV-2 specific humoral and cellular immunity in Omicron-infected individuals.
Collapse
Affiliation(s)
- Zhizhong Tang
- Urology Surgery Department, Maoming People's Hospital, Maoming, 525000, People's Republic of China
| | - Pei Yu
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Qianfang Guo
- Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Institute of Microbiology, Guangdong Provincial Center for Disease Control and Prevention, Guangdong, 511430, People's Republic of China
| | - Mingxiao Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Yu Lei
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Lei Zhou
- Department Of Pathology Laboratory, Maoming People's Hospital, Maoming, 525000, People's Republic of China
| | - Weikang Mai
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Lu Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Min Deng
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Weiya Kong
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Chuanying Niu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, People's Republic of China
| | - Xiaoli Xiong
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, People's Republic of China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, 510005, People's Republic of China
| | - Wenrui Li
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, People's Republic of China
| | - Chunbo Chen
- Intensive Care Unit Department, Maoming People's Hospital, Maoming, 525000, People's Republic of China
| | - Changchun Lai
- Clinical Laboratory Medicine Department, Maoming People's Hospital, Maoming, 525000, People's Republic of China.
| | - Qian Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China.
| | - Baisheng Li
- Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Institute of Microbiology, Guangdong Provincial Center for Disease Control and Prevention, Guangdong, 511430, People's Republic of China.
| | - Tianxing Ji
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China.
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511495, People's Republic of China.
| |
Collapse
|
19
|
Tsai DY, Wang CH, Schiro PG, Chen N, Tseng JY. Tracking B Cell Memory to SARS-CoV-2 Using Rare Cell Analysis System. Vaccines (Basel) 2023; 11:vaccines11040735. [PMID: 37112647 PMCID: PMC10145117 DOI: 10.3390/vaccines11040735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/14/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Rapid mutations within SARS-CoV-2 are driving immune escape, highlighting the need for in-depth and routine analysis of memory B cells (MBCs) to complement the important but limited information from neutralizing antibody (nAb) studies. In this study, we collected plasma samples and peripheral blood mononuclear cells (PBMCs) from 35 subjects and studied the nAb titers and the number of antigen-specific memory B cells at designated time points before and after vaccination. We developed an assay to use the MiSelect R II System with a single-use microfluidic chip to directly detect the number of spike-receptor-binding domain (RBD)-specific MBCs in PBMCs. Our results show that the number of spike-RBD-specific MBCs detected by the MiSelect R II System is highly correlated with the level of nAbs secreted by stimulated PBMCs, even 6 months after vaccination when nAbs were generally not present in plasma. We also found antigen-specific cells recognizing Omicron spike-RBD were present in PBMCs from booster vaccination of subjects, but with a high variability in the number of B cells. The MiSelect R II System provided a direct, automated, and quantitative method to isolate and analyze subsets of rare cells for tracking cellular immunity in the context of a rapidly mutating virus.
Collapse
Affiliation(s)
- Dong-Yan Tsai
- MiCareo Taiwan Co., Ltd., 5F, No. 69, Ln. 77, Xing Ai Rd., Neihu Dist., Taipei City 114, Taiwan
| | - Chun-Hung Wang
- MiCareo Taiwan Co., Ltd., 5F, No. 69, Ln. 77, Xing Ai Rd., Neihu Dist., Taipei City 114, Taiwan
| | - Perry G. Schiro
- MiCareo Taiwan Co., Ltd., 5F, No. 69, Ln. 77, Xing Ai Rd., Neihu Dist., Taipei City 114, Taiwan
| | - Nathan Chen
- Adimmune Corporation, No. 3, Sec.1, Tanxing Rd., Tanzi Dist., Taichung City 427, Taiwan
| | - Ju-Yu Tseng
- MiCareo Taiwan Co., Ltd., 5F, No. 69, Ln. 77, Xing Ai Rd., Neihu Dist., Taipei City 114, Taiwan
- Correspondence: ; Tel.: +886-2-27923976
| |
Collapse
|
20
|
Choi HL, Yang HR, Shin HG, Hwang K, Kim JW, Lee JH, Ryu T, Jung Y, Lee S. Generation and Next-Generation Sequencing-Based Characterization of a Large Human Combinatorial Antibody Library. Int J Mol Sci 2023; 24:ijms24066011. [PMID: 36983085 PMCID: PMC10057307 DOI: 10.3390/ijms24066011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/04/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Antibody phage display is a key technology for the discovery and development of target-specific monoclonal antibodies (mAbs) for use in research, diagnostics, and therapy. The construction of a high-quality antibody library, with larger and more diverse antibody repertoires, is essential for the successful development of phage display-derived mAbs. In this study, a large human combinatorial single-chain variable fragment library (1.5 × 1011 colonies) was constructed from Epstein-Barr virus-infected human peripheral blood mononuclear cells stimulated with a combination of two of the activators of human B cells, the Toll-like receptor 7/8 agonist R848 and interleukin-2. Next-generation sequencing analysis with approximately 1.9 × 106 and 2.7 × 106 full-length sequences of heavy chain variable (VH) and κ light chain variable (Vκ) domains, respectively, revealed that the library consists of unique VH (approximately 94%) and Vκ (approximately 91%) sequences with greater diversity than germline sequences. Lastly, multiple unique mAbs with high affinity and broad cross-species reactivity could be isolated from the library against two therapeutically relevant target antigens, validating the library quality. These findings suggest that the novel antibody library we have developed may be useful for the rapid development of target-specific phage display-derived recombinant human mAbs for use in therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Hye Lim Choi
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ha Rim Yang
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ha Gyeong Shin
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Kyusang Hwang
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ji Woong Kim
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ji Hyun Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Taehoon Ryu
- ATG Lifetech Inc., Seoul 08507, Republic of Korea
| | - Yushin Jung
- ATG Lifetech Inc., Seoul 08507, Republic of Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Department of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
21
|
Rouers A, Tay MZ, Ng LFP, Renia L. B-cell ELISpot assay to analyze human memory B cell and plasmablast responses specific to SARS-CoV-2 receptor-binding domain. STAR Protoc 2023; 4:102130. [PMID: 36853725 PMCID: PMC9910018 DOI: 10.1016/j.xpro.2023.102130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/19/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
B-cell ELISpot is an extremely sensitive assay based on the secretion of antibodies by B cells that requires the differentiation of B cells into antibody-secreting cells. Here, we describe the procedure to analyze both plasmablast (PB) and memory B cell (MBC) responses specific to SARS-CoV-2 receptor-binding domain (RBD) in the context of acute SARS-CoV-2 infection and vaccination. We detail steps for MBC stimulation, MBC and PB plating, detection, and counting of total IgG and RBD-specific spots. For complete details on the use and execution of this protocol, please refer to Tay et al. (2022).1.
Collapse
Affiliation(s)
- Angeline Rouers
- A∗STAR Infectious Diseases Labs (A∗STAR ID Labs), Agency for Science, Technology and Research (A∗STAR), 8A Biomedical Grove, Immunos #05-13, Singapore 138648, Singapore.
| | - Matthew Zirui Tay
- A∗STAR Infectious Diseases Labs (A∗STAR ID Labs), Agency for Science, Technology and Research (A∗STAR), 8A Biomedical Grove, Immunos #05-13, Singapore 138648, Singapore
| | - Lisa F P Ng
- A∗STAR Infectious Diseases Labs (A∗STAR ID Labs), Agency for Science, Technology and Research (A∗STAR), 8A Biomedical Grove, Immunos #05-13, Singapore 138648, Singapore; National Institute of Health Research, Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, UK; Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Laurent Renia
- A∗STAR Infectious Diseases Labs (A∗STAR ID Labs), Agency for Science, Technology and Research (A∗STAR), 8A Biomedical Grove, Immunos #05-13, Singapore 138648, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
22
|
Meron-Sudai S, Asato V, Adler A, Bialik A, Goren S, Ariel-Cohen O, Reizis A, Mulard LA, Phalipon A, Cohen D. A Shigella flexneri 2a synthetic glycan-based vaccine induces a long-lasting immune response in adults. NPJ Vaccines 2023; 8:35. [PMID: 36894570 PMCID: PMC9998260 DOI: 10.1038/s41541-023-00624-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/09/2023] [Indexed: 03/11/2023] Open
Abstract
Shigella is a leading cause of moderate to severe diarrhea worldwide and of diarrhea-associated deaths in children under 5 years of age in low-and middle-income countries. A vaccine against shigellosis is in high demand. SF2a-TT15, a synthetic carbohydrate-based conjugate vaccine candidate against Shigella flexneri 2a (SF2a) was found safe and strongly immunogenic in adult volunteers. Here, SF2a-TT15 at 10 µg oligosaccharide (OS) vaccine dose is shown to induce a sustained immune response in magnitude and functionality in the majority of volunteers followed up 2 and 3 years post-vaccination. High levels of either one of the humoral parameters as well as the number of specific-IgG memory B-cells determined 3 months after vaccination were good predictors of the durability of the immune response. This study is the first to examine the long-term durability of antibody functionality and memory B-cell response induced by a Shigella vaccine candidate.
Collapse
Affiliation(s)
- Shiri Meron-Sudai
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Valeria Asato
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Amos Adler
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel.,Clinical Microbiology Laboratory, Tel-Aviv Sourasky Medical Center, 6 Weizmann Street, Tel Aviv, 6423906, Israel
| | - Anya Bialik
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Sophy Goren
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Ortal Ariel-Cohen
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Arava Reizis
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Laurence A Mulard
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Unité de Chimie des Biomolécules, F-75015, Paris, France
| | - Armelle Phalipon
- Institut Pasteur, Innovation Lab. Vaccines, F-75015, Paris, France
| | - Dani Cohen
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel.
| |
Collapse
|
23
|
Yu P, Liu Z, Zhu Z, Yang J, Deng M, Chen M, Lai C, Kong W, Xiong S, Wan L, Mai W, Chen L, Lei Y, Khan SA, Ruan J, Kang A, Guo X, Zhou Q, Li W, Chen Z, Liang Y, Li P, Zhang L, Ji T. Omicron variants breakthrough infection elicited higher specific memory immunity than third dose booster in healthy vaccinees. Virol Sin 2023; 38:233-243. [PMID: 36603767 PMCID: PMC10176432 DOI: 10.1016/j.virs.2022.12.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
Homologous booster, heterologous booster, and Omicron variants breakthrough infection (OBI) could improve the humoral immunity against Omicron variants. Questions concerning about memory B cells (MBCs) and T cells immunity against Omicron variants, features of long-term immunity, after booster and OBI, needs to be explored. Here, comparative analysis demonstrate antibody and T cell immunity against ancestral strain, Delta and Omicron variants in Omicron breakthrough infected patients (OBIPs) are comparable to that in Ad5-nCoV boosted healthy volunteers (HVs), higher than that in inactivated vaccine (InV) boosted HVs. However, memory B cells (MBCs) immunity against Omicron variants was highest in OBIPs, followed by Ad5-nCoV boosted and InV boosted HVs. OBIPs and Ad5-nCoV boosted HVs have higher classical MBCs and activated MBCs, and lower naïve MBCs and atypical MBCs relative to both vaccine boosted HVs. Collectively, these data indicate Omicron breakthrough infection elicit higher MBCs and T cells against SARS-CoV-2 especially Omicron variants relative to homologous InV booster and heterologous Ad5-nCoV booster.
Collapse
Affiliation(s)
- Pei Yu
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Zijian Liu
- State Key Laboratories of Respiratory Diseases, Guangdong-Hong Kong-Macao Joint Laboratory of Infectious Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, China
| | - Zhuoqi Zhu
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China
| | - Jiaqing Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Min Deng
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Mingxiao Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Changchun Lai
- Clinical Laboratory Medicine Department, Maoming People's Hospital, Maoming, 525000, China
| | - Weiya Kong
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Shilong Xiong
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Li Wan
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Weikang Mai
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Lu Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Yu Lei
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Shahzad Akbar Khan
- Laboratory of Pathology, Department of Pathobiology, University of Poonch Rawalakot Azad Kashmir Pakistan 12350, Pakistan
| | - Jianfeng Ruan
- Hospital Infection-Control Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - An Kang
- Medical Examination Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Xuguang Guo
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510140, China
| | - Qiang Zhou
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Wenrui Li
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China
| | - Zheng Chen
- Kidney Transplant Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
| | - Yuemei Liang
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China.
| | - Pingchao Li
- State Key Laboratories of Respiratory Diseases, Guangdong-Hong Kong-Macao Joint Laboratory of Infectious Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, China.
| | - Lei Zhang
- Kidney Transplant Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China; Department of Organ Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, 510630, China.
| | - Tianxing Ji
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China; Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
24
|
Izadi N, Hauk PJ. Cellular assays to evaluate B-cell function. J Immunol Methods 2023; 512:113395. [PMID: 36470409 DOI: 10.1016/j.jim.2022.113395] [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: 08/02/2022] [Revised: 10/31/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Inborn errors of immunity (IEI) that present with recurrent infections are largely due to antibody (Ab) deficiencies. Therefore, assessment of the B-cell and Ab compartment is a major part of immunologic evaluation. Here we provide an overview about cellular assays used to study B-cell function and focus on lymphocyte proliferation assay (LPA), opsonophagocytic assay (OPA), and the Enzyme-linked Immunosorbent Spot Assay (ELISPOT) including clinical applications and limitations of these techniques. LPAs assess ex-vivo cell proliferation in response to various stimuli. Clinically available LPAs utilize peripheral blood mononuclear cells and mostly assess T-cell proliferation with pokeweed mitogen considered the most B-cell specific stimulus. In the research setting, isolating B cells or using more B-cell specific stimuli such as CD40L with IL-4/IL-21 or the TLR9 ligand CpG can more specifically capture the proliferative ability of B cells. OPAs are functional in-vitro killing assays used to evaluate the ability of IgG Ab to induce phagocytosis applied when assessing the potency of vaccine candidates or along with avidity assays to evaluate the quality of secreted IgG. The B-cell ELISPOT assesses Ab production at a cellular level and can characterize the Ab response of particular B-cell subtypes. It can be used in patients on IgG therapy by capturing specific Abs produced by individual B cells, which is not affected by exogenous IgG from plasma donors, and when assessing the vaccine response in patients on immunomodulatory drugs that can affect memory B-cell function. Emerging approaches that are only available in research settings are also briefly introduced.
Collapse
Affiliation(s)
- Neema Izadi
- Children's Hospital Los Angeles and Keck School of Medicine, USC, 4650 Sunset Blvd, Los Angeles, CA 90027, United States of America.
| | - Pia J Hauk
- Children's Hospital Colorado, Section Allergy/Immunology, 13123 E 16th Avenue, Aurora, CO 80045, United States of America
| |
Collapse
|
25
|
Mise-Omata S, Ikeda M, Takeshita M, Uwamino Y, Wakui M, Arai T, Yoshifuji A, Murano K, Siomi H, Nakagawara K, Ohyagi M, Ando M, Hasegawa N, Saya H, Murata M, Fukunaga K, Namkoong H, Lu X, Yamasaki S, Yoshimura A. Memory B Cells and Memory T Cells Induced by SARS-CoV-2 Booster Vaccination or Infection Show Different Dynamics and Responsiveness to the Omicron Variant. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2104-2113. [PMID: 36426984 DOI: 10.4049/jimmunol.2200525] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/21/2022] [Indexed: 01/04/2023]
Abstract
Although the immunological memory produced by BNT162b2 vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been well studied and established, further information using different racial cohorts is necessary to understand the overall immunological response to vaccination. We evaluated memory B and T cell responses to the severe acute respiratory syndrome coronavirus 2 spike protein before and after the third booster using a Japanese cohort. Although the Ab titer against the spike receptor-binding domain (RBD) decreased significantly 8 mo after the second vaccination, the number of memory B cells continued to increase, whereas the number of memory T cells decreased slowly. Memory B and T cells from unvaccinated infected patients showed similar kinetics. After the third vaccination, the Ab titer increased to the level of the second vaccination, and memory B cells increased at significantly higher levels before the booster, whereas memory T cells recovered close to the second vaccination levels. In memory T cells, the frequency of CXCR5+CXCR3+CCR6- circulating follicular Th1 was positively correlated with RBD-specific Ab-secreting B cells. For the response to variant RBDs, although 60-80% of memory B cells could bind to the omicron RBD, their avidity was low, whereas memory T cells show an equal response to the omicron spike. Thus, the persistent presence of memory B and T cells will quickly upregulate Ab production and T cell responses after omicron strain infection, which prevents severe illness and death due to coronavirus disease 2019.
Collapse
Affiliation(s)
- Setsuko Mise-Omata
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Mari Ikeda
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Masaru Takeshita
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yoshifumi Uwamino
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan.,Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan
| | - Masatoshi Wakui
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tomoko Arai
- Clinical Laboratory, Keio University Hospital, Tokyo, Japan
| | - Ayumi Yoshifuji
- Division of Nephrology, Department of Internal Medicine, Tokyo Saiseikai Central Hospital, Tokyo, Japan
| | - Kensaku Murano
- Department of Molecular Biology, Keio University School of Medicine, Tokyo, Japan
| | - Haruhiko Siomi
- Department of Molecular Biology, Keio University School of Medicine, Tokyo, Japan
| | - Kensuke Nakagawara
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan.,Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Ohyagi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Ando
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Naoki Hasegawa
- Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Mitsuru Murata
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ho Namkoong
- Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan
| | - Xiuyuan Lu
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan; and
| | - Sho Yamasaki
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan; and.,Department of Molecular Immunology, Research Institute Microbial Disease, Osaka University, Osaka, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
26
|
Noto A, Joo V, Mancarella A, Suffiotti M, Pellaton C, Fenwick C, Perreau M, Pantaleo G. CXCL12 and CXCL13 Cytokine Serum Levels Are Associated with the Magnitude and the Quality of SARS-CoV-2 Humoral Responses. Viruses 2022; 14:2665. [PMID: 36560669 PMCID: PMC9785906 DOI: 10.3390/v14122665] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
A better understanding of the immunological markers associated with long-lasting immune responses to SARS-CoV-2 infection is of paramount importance. In the present study, we characterized SARS-CoV-2-specific humoral responses in hospitalized (ICU and non-ICU) and non-hospitalized individuals at six months post-onset of symptoms (POS) (N = 95). We showed that the proportion of individuals with detectable anti-SARS-CoV-2 IgG or neutralizing (NAb) responses and the titers of antibodies were significantly reduced in non-hospitalized individuals, compared to ICU- or non-ICU-hospitalized individuals at 6 months POS. Interestingly, SARS-CoV-2-specific memory B cells persist at 6 months POS in both ICU and non-ICU patients and were enriched in cells harboring an activated and/or exhausted phenotype. The frequency/phenotype of SARS-CoV-2-specific memory B cells and the magnitude of IgG or NAb responses at 6 months POS correlated with the serum immune signature detected at patient admission. In particular, the serum levels of CXCL13, IL-1RA, and G-CSF directly correlated with the frequency of Spike-specific B cells and the magnitude of Spike-specific IgG or NAb, while the serum levels of CXCL12 showed an antagonizing effect. Our results indicate that the balance between CXCL12 and CXCL13 is an early marker associated with the magnitude and the quality of the SARS-CoV-2 humoral memory.
Collapse
Affiliation(s)
- Alessandra Noto
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Victor Joo
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Antonio Mancarella
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Madeleine Suffiotti
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Celine Pellaton
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
- Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
27
|
Wolf C, Köppert S, Becza N, Kuerten S, Kirchenbaum GA, Lehmann PV. Antibody Levels Poorly Reflect on the Frequency of Memory B Cells Generated following SARS-CoV-2, Seasonal Influenza, or EBV Infection. Cells 2022; 11:cells11223662. [PMID: 36429090 PMCID: PMC9688940 DOI: 10.3390/cells11223662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
The scope of immune monitoring is to define the existence, magnitude, and quality of immune mechanisms operational in a host. In clinical trials and praxis, the assessment of humoral immunity is commonly confined to measurements of serum antibody reactivity without accounting for the memory B cell potential. Relying on fundamentally different mechanisms, however, passive immunity conveyed by pre-existing antibodies needs to be distinguished from active B cell memory. Here, we tested whether, in healthy human individuals, the antibody titers to SARS-CoV-2, seasonal influenza, or Epstein-Barr virus antigens correlated with the frequency of recirculating memory B cells reactive with the respective antigens. Weak correlations were found. The data suggest that the assessment of humoral immunity by measurement of antibody levels does not reflect on memory B cell frequencies and thus an individual's potential to engage in an anamnestic antibody response against the same or an antigenically related virus. Direct monitoring of the antigen-reactive memory B cell compartment is both required and feasible towards that goal.
Collapse
Affiliation(s)
- Carla Wolf
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Sebastian Köppert
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Noémi Becza
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, 53115 Bonn, Germany
| | - Greg A. Kirchenbaum
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
| | - Paul V. Lehmann
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
- Correspondence: ; Tel.: +1-(216)-791-5084
| |
Collapse
|
28
|
Puente H, Díaz I, Arguello H, Mencía-Ares Ó, Gómez-García M, Pérez-Pérez L, Vega C, Cortey M, Martín M, Rubio P, Carvajal A. Characterization and cross-protection of experimental infections with SeCoV and two PEDV variants. Transbound Emerg Dis 2022; 69:3225-3237. [PMID: 35918058 DOI: 10.1111/tbed.14674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/13/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023]
Abstract
The aim of this study was to characterize the infection of weaned pigs with swine enteric coronavirus (SeCoV) - a chimeric virus most likely originated from a recombination event between porcine epidemic diarrhoea virus (PEDV) and transmissible gastroenteritis virus, or its mutant porcine respiratory coronavirus - and two PEDV G1b variants, including a recently described recombinant PEDV-SeCoV (rPEDV-SeCoV), as well as to determine the degree of cross-protection achieved against the rPEDV-SeCoV. For this purpose, forty-eight 4-week-old weaned pigs were randomly allocated into four groups of 12 animals. Piglets within each group were primary inoculated with one of the investigated viral strains (B: PEDV; C: SeCoV and D: rPEDV-SeCoV) or mock-inoculated (A), and exposed to rPEDV-SeCOV at day 20 post-infection; thus, group A was primary challenged (-/rPEDV-SeCoV), groups B and C were subjected to a heterologous re-challenge (PEDV/rPEDV-SeCoV and SeCoV/rPEDV-SeCoV, respectively), and group D to a homologous re-challenge (rPEDV-SeCoV/rPEDV-SeCoV), Clinical signs, viral shedding, microscopic lesions and specific humoral and cellular immune responses (IgG, IgA, neutralizing antibodies and IgA and IFN-γ-secreting cells) were monitored. After primo-infection, all three viral strains induced an undistinguishable mild-to-moderate clinical disease with diarrhoea as the main sign and villus shortening lesions in the small intestine. In homologous re-challenged pigs, no clinical signs or lesions were observed, and viral shedding was only detected in a single animal. This fact may be explained by the significant high level of rPEDV-SeCoV-specific neutralizing antibodies found in these pigs before the challenge. In contrast, prior exposure to a different PEDV G1b variant or SeCoV only provided partial cross-protection, allowing rPEDV-SeCoV replication and shedding in faeces.
Collapse
Affiliation(s)
- Héctor Puente
- Facultad de Veterinaria, Departamento de Sanidad Animal, Universidad de León, León, Spain
| | - Ivan Díaz
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Héctor Arguello
- Facultad de Veterinaria, Departamento de Sanidad Animal, Universidad de León, León, Spain.,INDEGSAL, Instituto de Desarrollo Ganadero, Universidad de León, León, Spain
| | - Óscar Mencía-Ares
- Facultad de Veterinaria, Departamento de Sanidad Animal, Universidad de León, León, Spain
| | - Manuel Gómez-García
- Facultad de Veterinaria, Departamento de Sanidad Animal, Universidad de León, León, Spain
| | - Lucía Pérez-Pérez
- Facultad de Veterinaria, Departamento de Sanidad Animal, Universidad de León, León, Spain
| | - Clara Vega
- Facultad de Veterinaria, Departamento de Sanidad Animal, Universidad de León, León, Spain
| | - Martí Cortey
- Facultat de Veterinària, Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Margarita Martín
- Facultat de Veterinària, Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Pedro Rubio
- Facultad de Veterinaria, Departamento de Sanidad Animal, Universidad de León, León, Spain.,INDEGSAL, Instituto de Desarrollo Ganadero, Universidad de León, León, Spain
| | - Ana Carvajal
- Facultad de Veterinaria, Departamento de Sanidad Animal, Universidad de León, León, Spain.,INDEGSAL, Instituto de Desarrollo Ganadero, Universidad de León, León, Spain
| |
Collapse
|
29
|
Jeewandara C, Aberathna IS, Pushpakumara PD, Kamaladasa A, Guruge D, Wijesinghe A, Gunasekera B, Tanussiya S, Kuruppu H, Ranasinghe T, Dayarathne S, Dissanayake O, Gamalath N, Ekanayake D, Jayamali J, Jayathilaka D, Dissanayake M, Madusanka D, Jayadas TT, Mudunkotuwa A, Somathilake G, Harvie M, Nimasha T, Danasekara S, Wijayamuni R, Schimanski L, Rijal P, Tan TK, Dong T, Townsend A, Ogg GS, Malavige GN. Immune responses to Sinopharm/BBIBP-CorV in individuals in Sri Lanka. Immunology 2022; 167:275-285. [PMID: 35758860 PMCID: PMC11495257 DOI: 10.1111/imm.13536] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 06/15/2022] [Indexed: 11/29/2022] Open
Abstract
As there are limited data of the immunogenicity of the Sinopharm/BBIBP-CorV in different populations, antibody responses against different SARS-CoV-2 variants of concern and T cell responses, we investigated the immunogenicity of the vaccine, in individuals in Sri Lanka. SARS-CoV-2-specific antibodies were measured in 282 individuals who were seronegative at baseline, and ACE2 receptor blocking antibodies, antibodies to the receptor-binding domain (RBD) of the wild-type (WT), alpha, beta and delta variants, ex vivo and cultured IFNγ ELISpot assays, intracellular cytokine secretion assays and B cell ELISpot assays were carried out in a sub cohort of the vaccinees at 4 and 6 weeks (2 weeks after the second dose). Ninety-five percent of the vaccinees seroconverted, although the seroconversion rates were significantly lower (p < 0.001) in individuals >60 years (93.3%) compared to those who were 20-39 years (98.9%); 81.25% had ACE2 receptor blocking antibodies at 6 weeks, and there was no difference in these antibody titres in vaccine sera compared to convalescent sera (p = 0.44). Vaccinees had significantly less (p < 0.0001) antibodies to the RBD of WT and alpha, although there was no difference in antibodies to the RBD of beta and delta compared to convalescent sera; 27.7% of 46.4% of vaccinees had ex vivo IFNγ and cultured ELISpot responses respectively, and IFNγ and CD107a responses were detected by flow cytometry. Sinopharm/BBIBP-CorV appeared to induce a similar level of antibody responses against ACE2 receptor, delta and beta as seen following natural infection.
Collapse
Affiliation(s)
- Chandima Jeewandara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Inoka Sepali Aberathna
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Pradeep Darshana Pushpakumara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Achala Kamaladasa
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | | | - Ayesha Wijesinghe
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Banuri Gunasekera
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Shyrar Tanussiya
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Heshan Kuruppu
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Thushali Ranasinghe
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Shashika Dayarathne
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Osanda Dissanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Nayanathara Gamalath
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Dinithi Ekanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Jeewantha Jayamali
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Deshni Jayathilaka
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Madushika Dissanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Deshan Madusanka
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Tibutius Thanesh Jayadas
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Anushika Mudunkotuwa
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Gayasha Somathilake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Michael Harvie
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Thashmi Nimasha
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Saubhagya Danasekara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | | | - Lisa Schimanski
- MRC Human Immunology UnitMRC Weatherall Institute of Molecular Medicine, University of OxfordOxfordUK
- Centre for Translational ImmunologyChinese Academy of Medical Sciences Oxford Institute, University of OxfordOxfordUK
| | - Pramila Rijal
- MRC Human Immunology UnitMRC Weatherall Institute of Molecular Medicine, University of OxfordOxfordUK
- Centre for Translational ImmunologyChinese Academy of Medical Sciences Oxford Institute, University of OxfordOxfordUK
| | - Tiong K. Tan
- MRC Human Immunology UnitMRC Weatherall Institute of Molecular Medicine, University of OxfordOxfordUK
- Centre for Translational ImmunologyChinese Academy of Medical Sciences Oxford Institute, University of OxfordOxfordUK
| | - Tao Dong
- MRC Human Immunology UnitMRC Weatherall Institute of Molecular Medicine, University of OxfordOxfordUK
- Centre for Translational ImmunologyChinese Academy of Medical Sciences Oxford Institute, University of OxfordOxfordUK
| | - Alain Townsend
- MRC Human Immunology UnitMRC Weatherall Institute of Molecular Medicine, University of OxfordOxfordUK
- Centre for Translational ImmunologyChinese Academy of Medical Sciences Oxford Institute, University of OxfordOxfordUK
| | - Graham S. Ogg
- MRC Human Immunology UnitMRC Weatherall Institute of Molecular Medicine, University of OxfordOxfordUK
- Centre for Translational ImmunologyChinese Academy of Medical Sciences Oxford Institute, University of OxfordOxfordUK
| | - Gathsaurie Neelika Malavige
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular MedicineUniversity of Sri JayewardenepuraNugegodaSri Lanka
- MRC Human Immunology UnitMRC Weatherall Institute of Molecular Medicine, University of OxfordOxfordUK
| |
Collapse
|
30
|
Novel CD81 Mutations in a Chinese Patient Led to IgA Nephropathy and Impaired BCR Signaling. J Clin Immunol 2022; 42:1672-1684. [PMID: 35849269 DOI: 10.1007/s10875-022-01333-2] [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: 04/28/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022]
Abstract
PURPOSE CD81 deficiency is an extremely rare primary immunodeficiency disease characterized by severe and recurrent infections, IgA-related nephropathy, and profound hypogammaglobulinemia. Only one patient has been reported so far, and the pathogenesis remains unclear. Here, we identified a new case of CD81 deficiency and described its pathogenesis. METHODS We analyzed the clinical, genetic, and immunological features of the patient with CD81 deficiency, and explored the pathogenesis of her antibody deficiencies. RESULTS The major manifestation of this patient was unexpectedly not recurrent infections but IgA nephropathy with aberrant serum galactose-deficient IgA1. Whole-exome sequencing revealed novel biallelic mutations in CD81 gene that abolished the surface expression of CD81. B cells from the patient lack membrane CD19 and showed reduced switched memory B cells and transitional B cells. Decreased expression of key molecules pY and pBTK in BCR signaling were demonstrated by confocal microscopy. RNA sequencing revealed that genes associated with BCR signaling and immunoglobulins were downregulated in CD81-deficient B cells. In addition, the patient showed increased frequency of T follicular helper cells that biased to Th1-like subsets. CONCLUSION We reported the second patient with CD81 deficiency in the world and illustrated aberrant BCR signaling in the patient, therefore helping to unravel the mechanism of antibody deficiency in CD81-deficient patients.
Collapse
|
31
|
Hoffmann-Veltung H, Anabire NG, Ofori MF, Janhmatz P, Ahlborg N, Hviid L, Quintana MDP. Analysis of allelic cross-reactivity of monoclonal IgG antibodies by a multiplexed reverse FluoroSpot assay. eLife 2022; 11:e79245. [PMID: 35838346 PMCID: PMC9286747 DOI: 10.7554/elife.79245] [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: 04/05/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
The issue of antibody cross-reactivity is of central importance in immunology, and not least in protective immunity to Plasmodium falciparum malaria, where key antigens show substantial allelic variation (polymorphism). However, serological analysis often does not allow the distinction between true cross-reactivity (one antibody recognizing multiple antigen variants) and apparent cross-reactivity (presence of multiple variant-specific antibodies), as it requires analysis at the single B-cell/monoclonal antibody level. ELISpot is an assay that enables that, and a recently developed multiplexed variant of ELISpot (FluoroSpot) facilitates simultaneous assessment of B-cell/antibody reactivity to several different antigens. In this study, we present a further enhancement of this assay that makes direct analysis of monoclonal antibody-level cross-reactivity with allelic variants feasible. Using VAR2CSA-type PfEMP1-a notoriously polymorphic antigen involved in the pathogenesis of placental malaria-as a model, we demonstrate the robustness of the assay and its applicability to analysis of true cross-reactivity of monoclonal VAR2CSA-specific antibodies in naturally exposed individuals. The assay is adaptable to the analysis of other polymorphic antigens, rendering it a powerful tool in studies of immunity to malaria and many other diseases.
Collapse
Affiliation(s)
- Henriette Hoffmann-Veltung
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Nsoh Godwin Anabire
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, University of GhanaAccraGhana
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of GhanaAccraGhana
| | - Michael Fokuo Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of GhanaAccraGhana
| | | | - Niklas Ahlborg
- Mabtech ABNacka StrandSweden
- Department of Molecular Biosciences, The Wenner-Gren InstituteStockholmSweden
| | - Lars Hviid
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
- Department of Infectious Diseases, RigshospitaletCopenhagenDenmark
| | - Maria del Pilar Quintana
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| |
Collapse
|
32
|
Ansari A, Sachan S, Jit BP, Sharma A, Coshic P, Sette A, Weiskopf D, Gupta N. An efficient immunoassay for the B cell help function of SARS-CoV-2-specific memory CD4 + T cells. CELL REPORTS METHODS 2022; 2:100224. [PMID: 35571764 PMCID: PMC9085463 DOI: 10.1016/j.crmeth.2022.100224] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/27/2021] [Accepted: 04/28/2022] [Indexed: 04/30/2023]
Abstract
The B cell "help" function of CD4+ T cells is an important mechanism of adaptive immunity. Here, we describe improved antigen-specific T-B cocultures for quantitative measurement of T cell-dependent B cell responses, with as few as ∼90 T cells. Utilizing M. tuberculosis (Mtb), we show that early priming and activation of CD4+ T cells is important for productive interaction between T and B cells and that similar effects are achieved by supplementing cocultures with monocytes. We find that monocytes promote survivability of B cells via BAFF and stem cell growth factor (SCGF)/C-type lectin domain family 11 member A (CLEC11A), but this alone does not fully recapitulate the effects of monocyte supplementation. Importantly, we demonstrate improved activation and immunological output of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific memory CD4+ T-B cell cocultures with the inclusion of monocytes. This method may therefore provide a more sensitive assay to evaluate the B cell help quality of memory CD4+ T cells, for example, after vaccination or natural infection.
Collapse
Affiliation(s)
- Asgar Ansari
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Shilpa Sachan
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Bimal Prasad Jit
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Poonam Coshic
- Department of Transfusion Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Nimesh Gupta
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| |
Collapse
|
33
|
Versteegen P, Barkoff AM, Valente Pinto M, van de Kasteele J, Knuutila A, Bibi S, de Rond L, Teräsjärvi J, Sanders K, de Zeeuw-Brouwer ML, Luoto R, ten Hulscher H, Clutterbuck EA, Sanders EAM, Mertsola J, Berbers GAM, He Q, Kelly DF, Buisman AM, PERISCOPE Consortium. Memory B Cell Activation Induced by Pertussis Booster Vaccination in Four Age Groups of Three Countries. Front Immunol 2022; 13:864674. [PMID: 35677044 PMCID: PMC9168128 DOI: 10.3389/fimmu.2022.864674] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundImmunogenicity of acellular pertussis (aP) vaccines is conventionally assessed by measuring antibody responses but antibody concentrations wane quickly after vaccination. Memory B cells, however, are critical in sustaining long-term protection and therefore may be an important factor when assessing pertussis immunity after vaccination.AimWe studied pertussis specific memory B cell (re)activation induced by an aP booster vaccination in four different age groups within three countries.Materials and methodsFrom a phase IV longitudinal interventional study, 268 participants across Finland, the Netherlands and the United Kingdom were included and received a 3-component pertussis booster vaccine: children (7-10y, n=53), adolescents (11-15y, n=66), young adults (20-34y, n=74), and older adults (60-70y, n=75). Memory B cells at baseline, day 28, and 1 year post-vaccination were measured by a pertussis toxin (Ptx), filamentous haemagglutinin (FHA), and pertactin (Prn) specific ELISpot assay. Antibody results measured previously were available for comparison. Furthermore, study participants were distributed into groups based on their baseline memory B cell frequencies, vaccine responses were monitored between these groups.ResultsGeometric mean (GM) memory B cell frequencies for pertussis antigens at baseline were low. At 28 days post-vaccination, these frequencies increased within each age group and were still elevated one year post-booster compared to baseline. Highest frequencies at day 28 were found within adolescents (GM: 5, 21, and 13, for Ptx, FHA and Prn, respectively) and lowest within older adults (GM: 2, 9, and 3, respectively). Moderate to strong correlations between memory B cell frequencies at day 28 and antibody concentrations at day 28 and 1 year were observed for Prn. Memory B cell frequencies > 1 per 100,000 PBMCs at baseline were associated with significantly higher memory responses after 28 days and 1 year.ConclusionsAn aP booster vaccine (re)activated memory B cells in all age groups. Still elevated memory B cell frequencies after one year indicates enhanced immunological memory. However, antigen specific memory B cell activation seems weaker in older adults, which might reflect immunosenescence. Furthermore, the presence of circulating memory B cells at baseline positively affects memory B cell responses. This study was registered at www.clinicaltrialsregister.eu: No. 2016-003678-42.
Collapse
Affiliation(s)
- Pauline Versteegen
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | - Alex-Mikael Barkoff
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Marta Valente Pinto
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Jan van de Kasteele
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | - Aapo Knuutila
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Sagida Bibi
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Lia de Rond
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | - Johanna Teräsjärvi
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Katherine Sanders
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Mary-lène de Zeeuw-Brouwer
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | - Raakel Luoto
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Hinke ten Hulscher
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | | | - Elisabeth A. M. Sanders
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital, Utrecht, Netherlands
| | - Jussi Mertsola
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Guy A. M. Berbers
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | - Qiushui He
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Dominic F. Kelly
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Anne-Marie Buisman
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
- *Correspondence: Anne-Marie Buisman,
| | | |
Collapse
|
34
|
Persistence of Virus-Specific Antibody after Depletion of Memory B Cells. J Virol 2022; 96:e0002622. [DOI: 10.1128/jvi.00026-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Following vaccination or primary virus infection, virus-specific antibodies provide the first line of defense against reinfection. Plasma cells residing in the bone marrow constitutively secrete antibodies, are long-lived, and can thus maintain serum antibody levels over extended periods of time in the absence of antigen.
Collapse
|
35
|
Liu Y, Zeng Q, Deng C, Li M, Li L, Liu D, Liu M, Ruan X, Mei J, Mo R, Zhou Q, Liu M, Peng S, Wang J, Zhang H, Xiao H. Robust induction of B cell and T cell responses by a third dose of inactivated SARS-CoV-2 vaccine. Cell Discov 2022; 8:10. [PMID: 35102140 PMCID: PMC8803973 DOI: 10.1038/s41421-022-00373-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022] Open
Abstract
SARS-CoV-2 inactivated vaccines have shown remarkable efficacy in clinical trials, especially in reducing severe illness and casualty. However, the waning of humoral immunity over time has raised concern over the durability of immune memory following vaccination. Thus, we conducted a nonrandomized trial among the healthcare workers (HCWs) to investigate the long-term sustainability of SARS-CoV-2-specific B cells and T cells stimulated by inactivated vaccines and the potential need for a third booster dose. Although neutralizing antibodies elicited by the standard two-dose vaccination schedule dropped from a peak of 29.3 arbitrary units (AU)/mL to 8.8 AU/mL 5 months after the second vaccination, spike-specific memory B and T cells were still detectable, forming the basis for a quick recall response. As expected, the faded humoral immune response was vigorously elevated to 63.6 AU/mL by 7.2 folds 1 week after the third dose along with abundant spike-specific circulating follicular helper T cells in parallel. Meanwhile, spike-specific CD4+ and CD8+ T cells were also robustly elevated by 5.9 and 2.7 folds respectively. Robust expansion of memory pools by the third dose potentiated greater durability of protective immune responses. Another key finding in this trial was that HCWs with low serological response to two doses were not truly "non-responders" but fully equipped with immune memory that could be quickly recalled by a third dose even 5 months after the second vaccination. Collectively, these data provide insights into the generation of long-term immunological memory by the inactivated vaccine, which could be rapidly recalled and further boosted by a third dose.
Collapse
Affiliation(s)
- Yihao Liu
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Clinical Trials Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qin Zeng
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Caiguanxi Deng
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mengyuan Li
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liubing Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dayue Liu
- Department of Medical Affairs, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ming Liu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xinyuan Ruan
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jie Mei
- Clinical Trials Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ruohui Mo
- Clinical Trials Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qian Zhou
- Clinical Trials Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Min Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Sui Peng
- Clinical Trials Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ji Wang
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Hui Zhang
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Haipeng Xiao
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
36
|
Winklmeier S, Eisenhut K, Taskin D, Rübsamen H, Gerhards R, Schneider C, Wratil PR, Stern M, Eichhorn P, Keppler OT, Klein M, Mader S, Kümpfel T, Meinl E. Persistence of functional memory B cells recognizing SARS-CoV-2 variants despite loss of specific IgG. iScience 2022; 25:103659. [PMID: 34957380 PMCID: PMC8686444 DOI: 10.1016/j.isci.2021.103659] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/17/2021] [Accepted: 12/15/2021] [Indexed: 01/22/2023] Open
Abstract
Although some COVID-19 patients maintain SARS-CoV-2-specific serum immunoglobulin G (IgG) for more than 6 months postinfection, others eventually lose IgG levels. We assessed the persistence of SARS-CoV-2-specific B cells in 17 patients, 5 of whom had lost specific IgGs after 5-8 months. Differentiation of blood-derived B cells in vitro revealed persistent SARS-CoV-2-specific IgG B cells in all patients, whereas IgA B cells were maintained in 11. Antibodies derived from cultured B cells blocked binding of viral receptor-binding domain (RBD) to the cellular receptor ACE-2, had neutralizing activity to authentic virus, and recognized the RBD of the variant of concern Alpha similarly to the wild type, whereas reactivity to Beta and Gamma were decreased. Thus, differentiation of memory B cells could be more sensitive for detecting previous infection than measuring serum antibodies. Understanding the persistence of SARS-CoV-2-specific B cells even in the absence of specific serum IgG will help to promote long-term immunity.
Collapse
Affiliation(s)
- Stephan Winklmeier
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Katharina Eisenhut
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Damla Taskin
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Heike Rübsamen
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Ramona Gerhards
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Celine Schneider
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Paul R. Wratil
- Max von Pettenkofer Institute & GeneCenter, Virology, LMU Munich, 80336 Munich, Germany
| | - Marcel Stern
- Max von Pettenkofer Institute & GeneCenter, Virology, LMU Munich, 80336 Munich, Germany
| | - Peter Eichhorn
- Institute of Laboratory Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Oliver T. Keppler
- Max von Pettenkofer Institute & GeneCenter, Virology, LMU Munich, 80336 Munich, Germany
| | - Matthias Klein
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Simone Mader
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
- Corresponding author
| |
Collapse
|
37
|
Jeffery-Smith A, Burton AR, Lens S, Rees-Spear C, Davies J, Patel M, Gopal R, Muir L, Aiano F, Doores KJ, Chow JY, Ladhani SN, Zambon M, McCoy LE, Maini MK. SARS-CoV-2-specific memory B cells can persist in the elderly who have lost detectable neutralizing antibodies. J Clin Invest 2022; 132:e152042. [PMID: 34843448 PMCID: PMC8759779 DOI: 10.1172/jci152042] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/24/2021] [Indexed: 11/24/2022] Open
Abstract
Memory B cells (MBCs) can provide a recall response able to supplement waning antibodies (Abs) with an affinity-matured response better able to neutralize variant viruses. We studied a cohort of elderly care home residents and younger staff (median age of 87 years and 56 years, respectively), who had survived COVID-19 outbreaks with only mild or asymptomatic infection. The cohort was selected because of its high proportion of individuals who had lost neutralizing antibodies (nAbs), thus allowing us to specifically investigate the reserve immunity from SARS-CoV-2-specific MBCs in this setting. Class-switched spike and receptor-binding domain (RBD) tetramer-binding MBCs persisted 5 months after mild or asymptomatic SARS-CoV-2 infection, irrespective of age. The majority of spike- and RBD-specific MBCs had a classical phenotype, but we found that activated MBCs, indicating possible ongoing antigenic stimulation or inflammation, were expanded in the elderly group. Spike- and RBD-specific MBCs remained detectable in the majority of individuals who had lost nAbs, although at lower frequencies and with a reduced IgG/IgA isotype ratio. Functional spike-, S1 subunit of the spike protein- (S1-), and RBD-specific recall was also detectable by enzyme-linked immune absorbent spot (ELISPOT) assay in some individuals who had lost nAbs, but was significantly impaired in the elderly. Our findings demonstrate that a reserve of SARS-CoV-2-specific MBCs persists beyond the loss of nAbs but highlight the need for careful monitoring of functional defects in spike- and RBD-specific B cell immunity in the elderly.
Collapse
Affiliation(s)
- Anna Jeffery-Smith
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
- Virus Reference Department, Public Health England (now called UK Health Security Agency [UKHSA]), London, United Kingdom
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Alice R. Burton
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Sabela Lens
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Chloe Rees-Spear
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Jessica Davies
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Monika Patel
- Virus Reference Department, Public Health England (now called UK Health Security Agency [UKHSA]), London, United Kingdom
| | - Robin Gopal
- Virus Reference Department, Public Health England (now called UK Health Security Agency [UKHSA]), London, United Kingdom
| | - Luke Muir
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Felicity Aiano
- Immunisation and Countermeasures Division, Public Health England (now called UKHSA), London, United Kingdom
| | - Katie J. Doores
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - J. Yimmy Chow
- London Coronavirus Response Cell, Public Health England (now called UKHSA), London, United Kingdom
| | - Shamez N. Ladhani
- Immunisation and Countermeasures Division, Public Health England (now called UKHSA), London, United Kingdom
| | - Maria Zambon
- Virus Reference Department, Public Health England (now called UK Health Security Agency [UKHSA]), London, United Kingdom
| | - Laura E. McCoy
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Mala K. Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| |
Collapse
|
38
|
Abstract
The enzyme-linked immunospot (ELISpot) is a highly sensitive immunoassay that measures the frequency of cytokine-secreting cells at the single-cell level. The secreted molecules are detected by using a detection antibody system similar to that used in the enzyme-linked immunosorbent assay (ELISA). The ELISpot assay is carried out in a 96-well plate and an automated ELISpot reader is used for analysis. The assay is easy to perform, robust and allows rapid analysis of a large number of samples and is not limited to measurement of cytokines; it is suitable for almost any secreted protein where single-cell analysis is of interest.
Collapse
|
39
|
Li Y, Mateu E, Díaz I. Impact of Cryopreservation on Viability, Phenotype, and Functionality of Porcine PBMC. Front Immunol 2021; 12:765667. [PMID: 34912338 PMCID: PMC8666977 DOI: 10.3389/fimmu.2021.765667] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
The use of frozen peripheral blood mononuclear cells (PBMC) is common in immunological studies. The impact of freezing PBMC has been assessed using human and mice cells, but little information is available regarding domestic animals. In the present study, the phenotype and functionality of frozen porcine PBMC were examined. In a preliminary experiment, three freezing media: fetal bovine serum plus 10% dimethyl sulfoxide, PSC cryopreservation kit, and Cryostor CS10, were compared regarding the preservation of cell viability and the response of PBMC to mitogens after thawing. After being stored one month in liquid nitrogen, cell viability was above 89% for all freezing media. The ELISPOT IFN-gamma (IFN-γ) results in response to PHA and of IgG ELISPOT in response to R848+IL-2 were similar to those obtained using fresh PBMC. In the second set of experiments, PBMC were obtained from five pigs vaccinated against Porcine reproductive and respiratory syndrome virus (PRRSV) and then frozen using Cryostor CS10. Recovered cells were phenotyped by flow cytometry using anti-CD3, CD4, CD8, and CD21 antibodies and were used to assess the PRRSV-specific responses in a proliferation experiment, an IFN-γ ELISPOT, and an IgG ELISPOT, and compared to the results obtained with fresh cells. The antigen-specific responses of frozen cells were significantly (p<0.05) impaired in the proliferation assay, particularly for CD4/CD8 double-positive T-cells and for CD21+ cells. Freezing resulted in decreased proliferation when Con A, but not PHA, was used. In ELISPOT, cryopreservation resulted in a decreased frequency of IFN-γ-secreting cells in response to PRRSV (p<0.05) but the response to PHA was not affected. No differences were observed in the IgG ELISPOT after polyclonal activation. Taken together, cryopreservation of porcine PBMC had a significant impact on the magnitude of recall antigen responses and therefore, it may affect the response of effector/memory cells but seems not to have a major impact on naïve T-cells. These results may help to the better use of frozen porcine PBMC, and to the interpretation of the results obtained from them.
Collapse
Affiliation(s)
- Yanli Li
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Enric Mateu
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,Centre de Recerca en Sanitat Animal, Institut de Recerca en Tecnologies Agroalimentàries (IRTA-CReSA), Bellaterra, Spain.,World Organisation for Animal Health (OIE) Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Bellaterra, Spain
| | - Ivan Díaz
- Centre de Recerca en Sanitat Animal, Institut de Recerca en Tecnologies Agroalimentàries (IRTA-CReSA), Bellaterra, Spain.,World Organisation for Animal Health (OIE) Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Bellaterra, Spain
| |
Collapse
|
40
|
Goel RR, Painter MM, Apostolidis SA, Mathew D, Meng W, Rosenfeld AM, Lundgreen KA, Reynaldi A, Khoury DS, Pattekar A, Gouma S, Kuri-Cervantes L, Hicks P, Dysinger S, Hicks A, Sharma H, Herring S, Korte S, Baxter AE, Oldridge DA, Giles JR, Weirick ME, McAllister CM, Awofolaju M, Tanenbaum N, Drapeau EM, Dougherty J, Long S, D’Andrea K, Hamilton JT, McLaughlin M, Williams JC, Adamski S, Kuthuru O, The UPenn COVID Processing Unit ‡, Frank I, Betts MR, Vella LA, Grifoni A, Weiskopf D, Sette A, Hensley SE, Davenport MP, Bates P, Luning Prak ET, Greenplate AR, Wherry EJ. mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science 2021; 374:abm0829. [PMID: 34648302 PMCID: PMC9284784 DOI: 10.1126/science.abm0829] [Citation(s) in RCA: 623] [Impact Index Per Article: 155.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/10/2021] [Indexed: 12/13/2022]
Abstract
The durability of immune memory after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) messenger RNA (mRNA) vaccination remains unclear. In this study, we longitudinally profiled vaccine responses in SARS-CoV-2–naïve and –recovered individuals for 6 months after vaccination. Antibodies declined from peak levels but remained detectable in most subjects at 6 months. By contrast, mRNA vaccines generated functional memory B cells that increased from 3 to 6 months postvaccination, with the majority of these cells cross-binding the Alpha, Beta, and Delta variants. mRNA vaccination further induced antigen-specific CD4+ and CD8+ T cells, and early CD4+ T cell responses correlated with long-term humoral immunity. Recall responses to vaccination in individuals with preexisting immunity primarily increased antibody levels without substantially altering antibody decay rates. Together, these findings demonstrate robust cellular immune memory to SARS-CoV-2 and its variants for at least 6 months after mRNA vaccination.
Collapse
Affiliation(s)
- Rishi R. Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mark M. Painter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sokratis A. Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Wenzhao Meng
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aaron M. Rosenfeld
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kendall A. Lundgreen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - David S. Khoury
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Ajinkya Pattekar
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Leticia Kuri-Cervantes
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Philip Hicks
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Dysinger
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Amanda Hicks
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Harsh Sharma
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Herring
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott Korte
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Amy E. Baxter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A. Oldridge
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Josephine R. Giles
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Madison E. Weirick
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Christopher M. McAllister
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Moses Awofolaju
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nicole Tanenbaum
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Elizabeth M. Drapeau
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jeanette Dougherty
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sherea Long
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kurt D’Andrea
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jacob T. Hamilton
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Maura McLaughlin
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Justine C. Williams
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sharon Adamski
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - The UPenn COVID Processing Unit‡
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Infectious Disease, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Infectious Disease, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Ian Frank
- Division of Infectious Disease, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael R. Betts
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Laura A. Vella
- Division of Infectious Disease, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Scott E. Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Paul Bates
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eline T. Luning Prak
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison R. Greenplate
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - E. John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
41
|
Safety and Immunogenicity of M2-Deficient, Single Replication, Live Influenza Vaccine (M2SR) in Adults. Vaccines (Basel) 2021; 9:vaccines9121388. [PMID: 34960134 PMCID: PMC8707871 DOI: 10.3390/vaccines9121388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/09/2021] [Accepted: 11/18/2021] [Indexed: 11/17/2022] Open
Abstract
M2SR (M2-deficient single replication) is an investigational live intranasal vaccine that protects against multiple influenza A subtypes in influenza-naïve and previously infected ferrets. We conducted a phase 1, first-in-human, randomized, dose-escalation, placebo-controlled study of M2SR safety and immunogenicity. Adult subjects received a single intranasal administration with either placebo or one of three M2SR dose levels (106, 107 or 108 tissue culture infectious dose (TCID50)) expressing hemagglutinin and neuraminidase from A/Brisbane/10/2007 (H3N2) (24 subjects per group). Subjects were evaluated for virus replication, local and systemic reactions, adverse events (AE), and immune responses post-vaccination. Infectious virus was not detected in nasal swabs from vaccinated subjects. At least one AE (most commonly mild nasal rhinorrhea/congestion) was reported among 29%, 58%, and 83% of M2SR subjects administered a low, medium or high dose, respectively, and among 46% of placebo subjects. No subject had fever or a severe reaction to the vaccine. Influenza-specific serum and mucosal antibody responses and B- and T-cell responses were significantly more frequent among vaccinated subjects vs. placebo recipients. The M2SR vaccine was safe and well tolerated and generated dose-dependent durable serum antibody responses against diverse H3N2 influenza strains. M2SR demonstrated a multi-faceted immune response in seronegative and seropositive subjects.
Collapse
|
42
|
Abstract
The FluoroSpot assay is a development of the highly sensitive enzyme-linked immunospot (ELISpot) assay which enables functional measurement of immunity at the single-cell level. Both assays are performed in a 96-well format and measures the frequency of analyte-secreting cells, in ELISpot usually limited to one analyte per well due to the use of enzymes and precipitating substrates for detection. FluoroSpot, performed in a similar way as ELISpot, overcomes this limitation by detecting each analyte with an assigned fluorophore instead of an enzyme. By using readers equipped with fluorophore-specific filters, cells producing single or multiple cytokines can be identified simultaneously in the same well. This greatly facilitates the analysis of functionally distinct subpopulations in heterogenous cell samples, for example, the frequency of polyfunctional T cells, suggested to be of importance in various disease states. FluoroSpot maintains the simplicity and sensitivity of the ELISpot while taking the assay a step further towards a multiplex analysis and an in-depth understanding of the quality of an immune response. We describe here a 96-well plate method to analyze cells that have secreted up to four different cytokines simultaneously (Four-color Fluorospot).
Collapse
|
43
|
Goel RR, Painter MM, Apostolidis SA, Mathew D, Meng W, Rosenfeld AM, Lundgreen KA, Reynaldi A, Khoury DS, Pattekar A, Gouma S, Kuri-Cervantes L, Hicks P, Dysinger S, Hicks A, Sharma H, Herring S, Korte S, Baxter AE, Oldridge DA, Giles JR, Weirick ME, McAllister CM, Awofolaju M, Tanenbaum N, Drapeau EM, Dougherty J, Long S, D'Andrea K, Hamilton JT, McLaughlin M, Williams JC, Adamski S, Kuthuru O, Frank I, Betts MR, Vella LA, Grifoni A, Weiskopf D, Sette A, Hensley SE, Davenport MP, Bates P, Luning Prak ET, Greenplate AR, Wherry EJ. mRNA Vaccination Induces Durable Immune Memory to SARS-CoV-2 with Continued Evolution to Variants of Concern. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.08.23.457229. [PMID: 34462751 PMCID: PMC8404899 DOI: 10.1101/2021.08.23.457229] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
SARS-CoV-2 mRNA vaccines have shown remarkable efficacy, especially in preventing severe illness and hospitalization. However, the emergence of several variants of concern and reports of declining antibody levels have raised uncertainty about the durability of immune memory following vaccination. In this study, we longitudinally profiled both antibody and cellular immune responses in SARS-CoV-2 naïve and recovered individuals from pre-vaccine baseline to 6 months post-mRNA vaccination. Antibody and neutralizing titers decayed from peak levels but remained detectable in all subjects at 6 months post-vaccination. Functional memory B cell responses, including those specific for the receptor binding domain (RBD) of the Alpha (B.1.1.7), Beta (B.1.351), and Delta (B.1.617.2) variants, were also efficiently generated by mRNA vaccination and continued to increase in frequency between 3 and 6 months post-vaccination. Notably, most memory B cells induced by mRNA vaccines were capable of cross-binding variants of concern, and B cell receptor sequencing revealed significantly more hypermutation in these RBD variant-binding clones compared to clones that exclusively bound wild-type RBD. Moreover, the percent of variant cross-binding memory B cells was higher in vaccinees than individuals who recovered from mild COVID-19. mRNA vaccination also generated antigen-specific CD8+ T cells and durable memory CD4+ T cells in most individuals, with early CD4+ T cell responses correlating with humoral immunity at later timepoints. These findings demonstrate robust, multi-component humoral and cellular immune memory to SARS-CoV-2 and current variants of concern for at least 6 months after mRNA vaccination. Finally, we observed that boosting of pre-existing immunity with mRNA vaccination in SARS-CoV-2 recovered individuals primarily increased antibody responses in the short-term without significantly altering antibody decay rates or long-term B and T cell memory. Together, this study provides insights into the generation and evolution of vaccine-induced immunity to SARS-CoV-2, including variants of concern, and has implications for future booster strategies.
Collapse
Affiliation(s)
- Rishi R Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Mark M Painter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Sokratis A Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Wenzhao Meng
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aaron M Rosenfeld
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kendall A Lundgreen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - David S Khoury
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Ajinkya Pattekar
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Leticia Kuri-Cervantes
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Philip Hicks
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Dysinger
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Amanda Hicks
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Harsh Sharma
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Sarah Herring
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Scott Korte
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Amy E Baxter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A Oldridge
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Josephine R Giles
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Madison E Weirick
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Christopher M McAllister
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Moses Awofolaju
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nicole Tanenbaum
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Elizabeth M Drapeau
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jeanette Dougherty
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sherea Long
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kurt D'Andrea
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jacob T Hamilton
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Maura McLaughlin
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Justine C Williams
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Sharon Adamski
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ian Frank
- Division of Infectious Disease, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael R Betts
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Laura A Vella
- Division of Infectious Disease, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Scott E Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Paul Bates
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eline T Luning Prak
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison R Greenplate
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - E John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
44
|
Saso A, Kampmann B, Roetynck S. Vaccine-Induced Cellular Immunity against Bordetella pertussis: Harnessing Lessons from Animal and Human Studies to Improve Design and Testing of Novel Pertussis Vaccines. Vaccines (Basel) 2021; 9:877. [PMID: 34452002 PMCID: PMC8402596 DOI: 10.3390/vaccines9080877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
Pertussis ('whooping cough') is a severe respiratory tract infection that primarily affects young children and unimmunised infants. Despite widespread vaccine coverage, it remains one of the least well-controlled vaccine-preventable diseases, with a recent resurgence even in highly vaccinated populations. Although the exact underlying reasons are still not clear, emerging evidence suggests that a key factor is the replacement of the whole-cell (wP) by the acellular pertussis (aP) vaccine, which is less reactogenic but may induce suboptimal and waning immunity. Differences between vaccines are hypothesised to be cell-mediated, with polarisation of Th1/Th2/Th17 responses determined by the composition of the pertussis vaccine given in infancy. Moreover, aP vaccines elicit strong antibody responses but fail to protect against nasal colonisation and/or transmission, in animal models, thereby potentially leading to inadequate herd immunity. Our review summarises current knowledge on vaccine-induced cellular immune responses, based on mucosal and systemic data collected within experimental animal and human vaccine studies. In addition, we describe key factors that may influence cell-mediated immunity and how antigen-specific responses are measured quantitatively and qualitatively, at both cellular and molecular levels. Finally, we discuss how we can harness this emerging knowledge and novel tools to inform the design and testing of the next generation of improved infant pertussis vaccines.
Collapse
Affiliation(s)
- Anja Saso
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Beate Kampmann
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Sophie Roetynck
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| |
Collapse
|
45
|
Köppert S, Wolf C, Becza N, Sautto GA, Franke F, Kuerten S, Ross TM, Lehmann PV, Kirchenbaum GA. Affinity Tag Coating Enables Reliable Detection of Antigen-Specific B Cells in Immunospot Assays. Cells 2021; 10:cells10081843. [PMID: 34440612 PMCID: PMC8394687 DOI: 10.3390/cells10081843] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/09/2021] [Accepted: 07/16/2021] [Indexed: 11/26/2022] Open
Abstract
Assessment of humoral immunity to SARS-CoV-2 and other infectious agents is typically restricted to detecting antigen-specific antibodies in the serum. Rarely does immune monitoring entail assessment of the memory B-cell compartment itself, although it is these cells that engage in secondary antibody responses capable of mediating immune protection when pre-existing antibodies fail to prevent re-infection. There are few techniques that are capable of detecting rare antigen-specific B cells while also providing information regarding their relative abundance, class/subclass usage and functional affinity. In theory, the ELISPOT/FluoroSpot (collectively ImmunoSpot) assay platform is ideally suited for antigen-specific B-cell assessments since it provides this information at single-cell resolution for individual antibody-secreting cells (ASC). Here, we tested the hypothesis that antigen-coating efficiency could be universally improved across a diverse set of viral antigens if the standard direct (non-specific, low affinity) antigen absorption to the membrane was substituted by high-affinity capture. Specifically, we report an enhancement in assay sensitivity and a reduction in required protein concentrations through the capture of recombinant proteins via their encoded hexahistidine (6XHis) affinity tag. Affinity tag antigen coating enabled detection of SARS-CoV-2 Spike receptor binding domain (RBD)-reactive ASC, and also significantly improved assay performance using additional control antigens. Collectively, establishment of a universal antigen-coating approach streamlines characterization of the memory B-cell compartment after SARS-CoV-2 infection or COVID-19 vaccinations, and facilitates high-throughput immune-monitoring efforts of large donor cohorts in general.
Collapse
Affiliation(s)
- Sebastian Köppert
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Carla Wolf
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Noémi Becza
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
| | - Giuseppe A. Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (G.A.S.); (T.M.R.)
| | - Fridolin Franke
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, 53115 Bonn, Germany
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (G.A.S.); (T.M.R.)
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Paul V. Lehmann
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
| | - Greg A. Kirchenbaum
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
- Correspondence: ; Tel.: +1-(216)-791-5084
| |
Collapse
|
46
|
Barkoff AM, Knuutila A, Mertsola J, He Q. Evaluation of Anti-PT Antibody Response after Pertussis Vaccination and Infection: The Importance of Both Quantity and Quality. Toxins (Basel) 2021; 13:toxins13080508. [PMID: 34437379 PMCID: PMC8402585 DOI: 10.3390/toxins13080508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/21/2022] Open
Abstract
Pertussis toxin (PT) is considered the main virulence factor causing whooping cough or pertussis. The protein is widely studied and its composition was revealed and sequenced already during the 1980s. The human immune system creates a good response against PT when measured in quantity. However, the serum anti-PT antibodies wane rapidly, and only a small amount of these antibodies are found a few years after vaccination/infection. Therefore, multiple approaches to study the functionality (quality) of these antibodies, e.g., avidity, neutralizing capacity, and epitope specificity, have been investigated. In addition, the long-term B cell memory (Bmem) to PT is crucial for good protection throughout life. In this review, we summarize the findings from functional PT antibody and Bmem studies. These results are discussed in line with the quantity of serum anti-PT antibodies. PT neutralizing antibodies and anti-PT antibodies with proper avidity are crucial for good protection against the disease, and certain epitopes have been identified to have multiple functions in the protection. Although PT-specific Bmem responses are detectable at least five years after vaccination, long-term surveillance is lacking. Variation of the natural boosting of circulating Bordetella pertussis in communities is an important confounding factor in these memory studies.
Collapse
Affiliation(s)
- Alex-Mikael Barkoff
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
| | - Aapo Knuutila
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
| | - Jussi Mertsola
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, 20520 Turku, Finland
| | - Qiushui He
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
- Correspondence: ; Tel.: +358-40-472-2255
| |
Collapse
|
47
|
Matsuda Y, Watanabe T, Li XK. Approaches for Controlling Antibody-Mediated Allograft Rejection Through Targeting B Cells. Front Immunol 2021; 12:682334. [PMID: 34276669 PMCID: PMC8282180 DOI: 10.3389/fimmu.2021.682334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/17/2021] [Indexed: 01/14/2023] Open
Abstract
Both acute and chronic antibody-mediated allograft rejection (AMR), which are directly mediated by B cells, remain difficult to treat. Long-lived plasma cells (LLPCs) in bone marrow (BM) play a crucial role in the production of the antibodies that induce AMR. However, LLPCs survive through a T cell-independent mechanism and resist conventional immunosuppressive therapy. Desensitization therapy is therefore performed, although it is accompanied by severe side effects and the pathological condition may be at an irreversible stage when these antibodies, which induce AMR development, are detected in the serum. In other words, AMR control requires the development of a diagnostic method that predicts its onset before LLPC differentiation and enables therapeutic intervention and the establishment of humoral immune monitoring methods providing more detailed information, including individual differences in the susceptibility to immunosuppressive agents and the pathological conditions. In this study, we reviewed recent studies related to the direct or indirect involvement of immunocompetent cells in the differentiation of naïve-B cells into LLPCs, the limitations of conventional methods, and the possible development of novel control methods in the context of AMR. This information will significantly contribute to the development of clinical applications for AMR and improve the prognosis of patients who undergo organ transplantation.
Collapse
Affiliation(s)
- Yoshiko Matsuda
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takeshi Watanabe
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
48
|
Ticha O, Klemm D, Moos L, Bekeredjian-Ding I. A cell-based in vitro assay for testing of immunological integrity of Tetanus toxoid vaccine antigen. NPJ Vaccines 2021; 6:88. [PMID: 34162886 PMCID: PMC8222253 DOI: 10.1038/s41541-021-00344-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 05/24/2021] [Indexed: 12/03/2022] Open
Abstract
Vaccines containing inactivated toxins confer protection by eliciting a neutralizing antibody response against bacterial toxins such as tetanus and diphtheria. At present, release of tetanus toxoid (TT) and diphtheria toxoid (DT)-containing vaccines relies on in vivo experiments showing the protective vaccine response. The aim of this study was to develop a reliable in vitro assay for TT vaccine antigen characterization with the potential of replacing in vivo potency experiments. To this end, we exploited that TT elicits a recall response in vaccinated donors: human peripheral blood mononuclear cells (PBMC) were stimulated with alum-adsorbed TT bulk antigen and low concentrations of TLR9 ligand; induction of TT-specific IgG was quantified via ELISpot after 5 days. Proof-of-concept was obtained using paired samples from donors before and after vaccination; anti-TT IgG was only detected in PBMC collected after booster vaccination; specificity was demonstrated with DT stimulation as control. Notably, when using PBMC from buffy coats, the specific response to TT was reproducible in 30% of cells; responsiveness correlated with higher numbers of switched memory B cells. Consecutive results showed that TT-specific IgG was also detectable when PBMC were stimulated with DTaP final vaccine product. Thus, the assay provides a viable means to test B-cell differentiation and induction of TT-specific IgG secretion using bulk antigen and final vaccine. However, prequalification of PBMC is required for reliable performance. Along with physicochemical and immunochemical methods, the functional assay could represent a complementary tool to replace in vivo potency assays in batch release of TT-containing vaccines.
Collapse
Affiliation(s)
- Olga Ticha
- Division of Microbiology, Paul-Ehrlich-Institut, Langen, Germany
| | - Dido Klemm
- Division of Microbiology, Paul-Ehrlich-Institut, Langen, Germany
| | - Lukas Moos
- Division of Microbiology, Paul-Ehrlich-Institut, Langen, Germany
| | | |
Collapse
|
49
|
DiSano KD, Gilli F, Pachner AR. Memory B Cells in Multiple Sclerosis: Emerging Players in Disease Pathogenesis. Front Immunol 2021; 12:676686. [PMID: 34168647 PMCID: PMC8217754 DOI: 10.3389/fimmu.2021.676686] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/11/2021] [Indexed: 11/25/2022] Open
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Once thought to be primarily driven by T cells, B cells are emerging as central players in MS immunopathogenesis. Interest in multiple B cell phenotypes in MS expanded following the efficacy of B cell-depleting agents targeting CD20 in relapsing-remitting MS and inflammatory primary progressive MS patients. Interestingly, these therapies primarily target non-antibody secreting cells. Emerging studies seek to explore B cell functions beyond antibody-mediated roles, including cytokine production, antigen presentation, and ectopic follicle-like aggregate formation. Importantly, memory B cells (Bmem) are rising as a key B cell phenotype to investigate in MS due to their antigen-experience, increased lifespan, and rapid response to stimulation. Bmem display diverse effector functions including cytokine production, antigen presentation, and serving as antigen-experienced precursors to antibody-secreting cells. In this review, we explore the cellular and molecular processes involved in Bmem development, Bmem phenotypes, and effector functions. We then examine how these concepts may be applied to the potential role(s) of Bmem in MS pathogenesis. We investigate Bmem both within the periphery and inside the CNS compartment, focusing on Bmem phenotypes and proposed functions in MS and its animal models. Finally, we review how current immunomodulatory therapies, including B cell-directed therapies and other immunomodulatory therapies, modify Bmem and how this knowledge may be harnessed to direct therapeutic strategies in MS.
Collapse
Affiliation(s)
- Krista D. DiSano
- Department of Neurology, Geisel School of Medicine & Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | | | | |
Collapse
|
50
|
Horta S, Neumann F, Yeh SH, Langseth CM, Kangro K, Breukers J, Madaboosi N, Geukens N, Vanhoorelbeke K, Nilsson M, Lammertyn J. Evaluation of Immuno-Rolling Circle Amplification for Multiplex Detection and Profiling of Antigen-Specific Antibody Isotypes. Anal Chem 2021; 93:6169-6177. [PMID: 33823582 DOI: 10.1021/acs.analchem.1c00172] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Antibody characterization is essential for understanding the immune system and development of diagnostics and therapeutics. Current technologies are mainly focusing on the detection of antigen-specific immunoglobulin G (IgG) using bulk singleplex measurements, which lack information on other isotypes and specificity of individual antibodies. Digital immunoassays based on nucleic acid amplification have demonstrated superior performance by allowing the detection of single molecules in a multiplex and sensitive manner. In this study, we demonstrate for the first time an immuno-rolling circle amplification (immuno-RCA) assay for the multiplex detection of three antigen-specific antibody isotypes (IgG, IgA, and IgM) and its integration with microengraving. To validate this approach, we used the autoimmune disease immune-mediated thrombotic thrombocytopenic purpura (iTTP) as the model disease with anti-ADAMTS13 autoantibodies as the diagnostic target molecules. To identify the anti-ADAMTS13 autoantibody isotypes, we designed a pool of three unique antibody-oligonucleotide conjugates for identification and subsequent amplification and visualization via RCA. To validate this approach, we first confirmed an assay specificity of >88% and a low limit of detection of 0.3 ng/mL in the spiked buffer. Subsequently, we performed a dilution series of an iTTP plasma sample for the multiplex detection of the three isotypes with higher sensitivity compared to an enzyme-linked immunosorbent assay. Finally, we demonstrated single-cell analysis of human B cells and hybridoma cells for the detection of secreted antibodies using microengraving and achieved a detection of 23.3 pg/mL secreted antibodies per hour. This approach could help to improve the understanding of antibody isotype distributions and their roles in various diseases.
Collapse
Affiliation(s)
- Sara Horta
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Etienne Sabbelaan 53, Kortrijk 8500, Belgium.,Department of Biosystems, Biosensors Group, KU Leuven, Willem De Croylaan 42, Heverlee B-3001, Belgium
| | - Felix Neumann
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23B, Solna 171 65, Sweden
| | - Shu-Hao Yeh
- Department of Biosystems, Biosensors Group, KU Leuven, Willem De Croylaan 42, Heverlee B-3001, Belgium
| | - Christoffer Mattsson Langseth
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23B, Solna 171 65, Sweden
| | - Kadri Kangro
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Etienne Sabbelaan 53, Kortrijk 8500, Belgium.,Icosagen Cell Factory OÜ, Kambja vald, Tartumaa 61713, Estonia
| | - Jolien Breukers
- Department of Biosystems, Biosensors Group, KU Leuven, Willem De Croylaan 42, Heverlee B-3001, Belgium
| | - Narayanan Madaboosi
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23B, Solna 171 65, Sweden
| | - Nick Geukens
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Herestraat 49, Leuven 3000, Belgium
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Etienne Sabbelaan 53, Kortrijk 8500, Belgium
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23B, Solna 171 65, Sweden
| | - Jeroen Lammertyn
- Department of Biosystems, Biosensors Group, KU Leuven, Willem De Croylaan 42, Heverlee B-3001, Belgium
| |
Collapse
|