1
|
Pourabbasi Ardekan A, Haghighi A, Mohammadi-Yeganeh S, Ghorbani-Bidkorpeh F, Kashefi S, Koochaki A, Movahedi S, Rahmani Y, Najafi Dastenaei A, Haji Molla Hoseini M. Evaluation of the Immunoadjuvant Effects of miR-155-Chitosan Polyplex on Leishmania major Infected Mice. Immunol Invest 2024:1-17. [PMID: 39569986 DOI: 10.1080/08820139.2024.2430695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
BACKGROUND MicroRNAs have gained attention as key immunomodulators, with miR-155 specifically shown in various studies to drive macrophage polarization toward the classical phenotype. This polarization is crucial, as classical macrophages play a well-recognized role in differentiating type-1 immune responses and resisting Leishmania infection. OBJECTIVE The present study aims to evaluate the anti-leishmanial immunoadjuvant effects of the miR-155 chitosan polyplex (miR-155 CP). METHODS The anti-leishmanial immunoadjuvant activity of miR-155 CP synthesized by the coacervation method was assessed against L. major (MRHO/IR/75/ER) by analyzing the infectivity rate on RAW 264.7 cells in vitro.MiR-155 CP as an adjuvant co-administrated with soluble Leishmania antigen (SLA) for immunization of BALB/c mice, then the challenge was performed by subcutaneous injection of 1 × 106 L. major promastigotes. Eight weeks following the challenge, lesion size, parasite load, cytokine assay, and nitric oxide production were evaluated. RESULTS The nanoparticles were produced with a size of 233.87 ± 8 nm and a zeta potential of + 22.6 ± 2 mV with good transfection efficiency. The mean infection index among pretreated cells with miR-155 CP (72±1.1) decreased significantly compared to the control group (420 ± 2.8). The parasite burden and the size of the lesions were significantly reduced in the immunized infected mice. Vaccination by miR-155 CP/SLA triggered the production of IFN-γ and NO and changed the cytokine profile of antigen-specific cells.Conclusion:The effectiveness of the SLA vaccine can be enhanced by including miR-155 CP as an adjuvant. SLA and miR-155 CP co-administration improve the type-1 immune response. This enhanced immune response helps prevent severe leishmaniasis.
Collapse
Affiliation(s)
- Azam Pourabbasi Ardekan
- Department of Parasitology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Haghighi
- Department of Parasitology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ghorbani-Bidkorpeh
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sarvenaz Kashefi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ameneh Koochaki
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Movahedi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yasamin Rahmani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mostafa Haji Molla Hoseini
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Chen Y, Ma T. Hematologic cancers and infections: how to detect infections in advance and determine the type? Front Cell Infect Microbiol 2024; 14:1476543. [PMID: 39559703 PMCID: PMC11570547 DOI: 10.3389/fcimb.2024.1476543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024] Open
Abstract
Infection is one of the leading causes of death in patients with hematologic cancers. Hematologic cancer patients with compromised immune systems are already susceptible to infections, which come on even more rapidly and are difficult to control after they develop neutrophil deficiencies from high-dose chemotherapy. After patients have developed an infection, the determination of the type of infection becomes a priority for clinicians. In this review, we summarize the biomarkers currently used for the prediction of infections in patients with hematologic cancers; procalcitonin, CD64, cytokines, and CD14 et al. can be used to determine bacterial infections, and (1-3)-β-D-glucan and galactomannan et al. can be used as a determination of fungal infections. We have also focused on the use of metagenomic next-generation sequencing in infections in patients with hematologic cancers, which has excellent clinical value in infection prediction and can detect microorganisms that cannot be detected by conventional testing methods such as blood cultures. Of course, we also focused on infection biomarkers that are not yet used in blood cancer patients but could be used as a future research direction, e.g., human neutrophil lipocalin, serum amyloid A, and heparin-binding protein et al. Finally, clinicians need to combine multiple infection biomarkers, the patient's clinical condition, local susceptibility to the type of infection, and many other factors to make a determination of the type of infection.
Collapse
Affiliation(s)
- Yan Chen
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Tao Ma
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
3
|
Liu H, Bao H, Zhao J, Zhu F, Zheng C. Establishment and verification of a prognostic immune cell signature-based model for breast cancer overall survival. Transl Cancer Res 2024; 13:5600-5615. [PMID: 39525032 PMCID: PMC11543049 DOI: 10.21037/tcr-24-1829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Background Breast cancer (BRCA) is a prevalent and aggressive disease. Despite various treatments being applied, a significant number of patients continue to experience unfavorable prognoses. Accurate prognosis prediction in BRCA is crucial for tailoring individualized treatment plans and improving patient outcomes. Recent studies have highlighted the significance of immune cell infiltration in the tumor microenvironment (TME), but predicting survival remains challenging due to the heterogeneity of BRCA. The aim of this study was thus to produce an immune cell signature-based framework capable of predicting the prognosis of patients with BRCA. Methods The GSE169246 dataset was from the Gene Expression Omnibus (GEO) database, comprising single-cell RNA sequencing (scRNA-seq) data from 95 individuals with BRCA. Seurat, principal component analysis (PCA), the unified matrix polynomial approach (UMAP) algorithm, and linear dimensionality reduction were used to determine the heterogeneity of T cells. Overlapping analysis of differentially expressed genes (DEGs), genes associated with prognosis, and T-cell pharmacodynamics-related genes were used to obtain the T-cell core pharmacodynamics-related genes. The dimensionality of the T-cell core pharmacodynamics-related genes was reduced employing the least absolute shrinkage and selection operator (LASSO) Cox regression model and the LASSO model. The prognostic model was built via a Cox analysis of the overall survival (OS) information. The clinical sample included 95 patients with BRCA who underwent surgical treatment from October 2018 to October 2021 at the Second Affiliated Hospital of Qiqihar Medical University. Patients were divided into a good prognosis group and a poor prognosis group based on their prognostic outcomes. The predictive value of tumor characteristics and immune responses was validated through correlation analysis, logistic regression analysis, and receiver operating characteristic (ROC) analysis. Results A group of 95 genes was used to establish a prognostic model. In the GEO clinical sample, with a high-risk group demonstrating shorter median survival times (2,447 vs. 6,498 days, P=4.733e-12). Area under the curve (AUC) values of 0.75, 0.75, and 0.72 were obtained for 2-, 4-, and 6-year OS predictions, respectively. Clinical validation found that the 6-year OS of the favorable prognosis group was significantly higher than that of the unfavorable prognosis group (92.06% vs. 65.62%; P=0.005). Poor prognosis was positively correlated with age, tumor size, B-cell level, and CTLA4 level and negatively correlated with tumor stage (T1/T2), lymph node metastasis stage (N0), clinical stage I-II, CD3+T-cell, CD4+T-cell, CD8+T-cell, neutrophil, lymphocyte, natural kill cell, TIGIT expression and OS. The combined model of clinical parameters had an AUC value of 0.898. Conclusions This study established a prognostic model that demonstrated excellent predictive value for OS of BRCA. The predictive model developed offers valuable insights into prognosis and treatment planning, emphasizing the importance of tumor characteristics and immune cell infiltration.
Collapse
Affiliation(s)
- Hailong Liu
- Department of Surgical Oncology, the Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Hongguang Bao
- Department of Surgical Oncology, the Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Jingying Zhao
- Department of Surgical Oncology, the Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Fangxu Zhu
- Department of Surgical Oncology, the Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Chunlei Zheng
- Department of Surgical Oncology, the Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
4
|
Dvorscek AR, McKenzie CI, Stäheli VC, Ding Z, White J, Fabb SA, Lim L, O'Donnell K, Pitt C, Christ D, Hill DL, Pouton CW, Burnett DL, Brink R, Robinson MJ, Tarlinton DM, Quast I. Conversion of vaccines from low to high immunogenicity by antibodies with epitope complementarity. Immunity 2024; 57:2433-2452.e7. [PMID: 39305904 DOI: 10.1016/j.immuni.2024.08.017] [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: 11/10/2023] [Revised: 05/06/2024] [Accepted: 08/27/2024] [Indexed: 10/11/2024]
Abstract
Existing antibodies (Abs) have varied effects on humoral immunity during subsequent infections. Here, we leveraged in vivo systems that allow precise control of antigen-specific Abs and B cells to examine the impact of Ab dose, affinity, and specificity in directing B cell activation and differentiation. Abs competing with the B cell receptor (BCR) epitope showed affinity-dependent suppression. By contrast, Abs targeting a complementary epitope, not overlapping with the BCR, shifted B cell differentiation toward Ab-secreting cells. Such Abs allowed for potent germinal center (GC) responses to otherwise poorly immunogenic sites by promoting antigen capture and presentation by low-affinity B cells. These mechanisms jointly diversified the B cell repertoire by facilitating the recruitment of high- and low-affinity B cells into Ab-secreting cell, GC, and memory B cell fates. Incorporation of small amounts of monoclonal Abs into protein- or mRNA-based vaccines enhanced immunogenicity and facilitated sustained immune responses, with implications for vaccine design and our understanding of protective immunity.
Collapse
Affiliation(s)
- Alexandra R Dvorscek
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Craig I McKenzie
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Vera C Stäheli
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Zhoujie Ding
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Jacqueline White
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Stewart A Fabb
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Leonard Lim
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Kristy O'Donnell
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Catherine Pitt
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Daniel Christ
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Danika L Hill
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Deborah L Burnett
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2010, Australia
| | - Robert Brink
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Marcus J Robinson
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - David M Tarlinton
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Isaak Quast
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia.
| |
Collapse
|
5
|
Hu T, Wang Y, Wang Y, Cui H, Zhang J, Chen H, Wu B, Hao S, Chu CC, Wu Y, Zeng W. Production and evaluation of three kinds of vaccines against largemouth bass virus, and DNA vaccines show great application prospects. FISH & SHELLFISH IMMUNOLOGY 2024; 153:109841. [PMID: 39173984 DOI: 10.1016/j.fsi.2024.109841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
Largemouth bass virus (LMBV) infections has resulted in high mortality and economic losses to the global largemouth bass industry and has seriously restricted the healthy development of the bass aquaculture industry. There are currently no antiviral therapies available for the control of this disease. In this study, we developed three types of vaccine against LMBV; whole virus inactivated vaccine (I), a subunit vaccine composed of the major viral capsid protein MCP (S) as well as an MCP DNA vaccine(D), These were employed using differing immunization and booster strategies spaced 2 weeks apart as follows: II, SS, DD and DS. We found that all vaccine groups induced humoral and cellular immune responses and protected largemouth bass from a lethal LMBV challenge to varying degrees and DD produced the best overall effect. Specifically, the levels of specific IgM in serum in all immunized groups were elevated and significantly higher than those in the control group. Moreover, the expression of humoral immunity (CD4 and IgM) and cellular immunity (MHCI-α) as well as cytokines (IL-1β) was increased, and the activity of immunity-related enzymes ACP, AKP, LZM, and T-SOD in the serum was significantly enhanced. In addition, the relative percent survival of fish following an LMBV lethal challenge 4 weeks after the initial immunizations were high for each group: DD(89.5 %),DS(63.2 %),SS(50 %) and II (44.7 %). These results indicated that the MCP DNA vaccine is the most suitable and promising vaccine candidate for the effective control of LMBV disease.
Collapse
Affiliation(s)
- Tianmei Hu
- School of Life Science and Engineering, Foshan University, Foshan, 528225, PR China
| | - Yaoda Wang
- School of Life Science and Engineering, Foshan University, Foshan, 528225, PR China
| | - Yuhui Wang
- School of Life Science and Engineering, Foshan University, Foshan, 528225, PR China
| | - Hongye Cui
- School of Life Science and Engineering, Foshan University, Foshan, 528225, PR China
| | - Jiping Zhang
- School of Life Science and Engineering, Foshan University, Foshan, 528225, PR China
| | - Haiyue Chen
- School of Life Science and Engineering, Foshan University, Foshan, 528225, PR China
| | - Baozhou Wu
- School of Life Science and Engineering, Foshan University, Foshan, 528225, PR China
| | - Shuguang Hao
- School of Life Science and Engineering, Foshan University, Foshan, 528225, PR China
| | - Chien Chi Chu
- School of Life Science and Engineering, Foshan University, Foshan, 528225, PR China
| | - Yali Wu
- Foshan Institute of Agricultural Sciences, Foshan, 528145, Guangdong, PR China
| | - Weiwei Zeng
- School of Life Science and Engineering, Foshan University, Foshan, 528225, PR China.
| |
Collapse
|
6
|
Ying B, Liang CY, Desai P, Scheaffer SM, Elbashir SM, Edwards DK, Thackray LB, Diamond MS. Ipsilateral or contralateral boosting of mice with mRNA vaccines confers equivalent immunity and protection against a SARS-CoV-2 Omicron strain. J Virol 2024; 98:e0057424. [PMID: 39194250 PMCID: PMC11406931 DOI: 10.1128/jvi.00574-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Boosting with mRNA vaccines encoding variant-matched spike proteins has been implemented to mitigate their reduced efficacy against emerging SARS-CoV-2 variants. Nonetheless, in humans, it remains unclear whether boosting in the ipsilateral or contralateral arm with respect to the priming doses impacts immunity and protection. Here, we boosted K18-hACE2 mice with either monovalent mRNA-1273 (Wuhan-1 spike) or bivalent mRNA-1273.214 (Wuhan-1 + BA.1 spike) vaccine in the ipsilateral or contralateral leg after a two-dose priming series with mRNA-1273. Boosting in the ipsilateral or contralateral leg elicited equivalent levels of serum IgG and neutralizing antibody responses against Wuhan-1 and BA.1. While contralateral boosting with mRNA vaccines resulted in the expansion of spike-specific B and T cells beyond the ipsilateral draining lymph node (DLN) to the contralateral DLN, administration of a third mRNA vaccine dose at either site resulted in similar levels of antigen-specific germinal center B cells, plasmablasts/plasma cells, T follicular helper cells, and CD8+ T cells in the DLNs and the spleen. Furthermore, ipsilateral and contralateral boosting with mRNA-1273 or mRNA-1273.214 vaccines conferred similar homologous or heterologous immune protection against SARS-CoV-2 BA.1 virus challenge with equivalent reductions in viral RNA and infectious virus in the nasal turbinates and lungs. Collectively, our data show limited differences in B and T cell immune responses after ipsilateral and contralateral site boosting by mRNA vaccines that do not substantively impact protection against an Omicron strain.IMPORTANCESequential boosting with mRNA vaccines has been an effective strategy to overcome waning immunity and neutralization escape by emerging SARS-CoV-2 variants. However, it remains unclear how the site of boosting relative to the primary vaccination series shapes optimal immune responses or breadth of protection against variants. In K18-hACE2 transgenic mice, we observed that intramuscular boosting with historical monovalent or variant-matched bivalent vaccines in the ipsilateral or contralateral limb elicited comparable levels of serum spike-specific antibody and antigen-specific B and T cell responses. Moreover, boosting on either side conferred equivalent protection against a SARS-CoV-2 Omicron challenge strain. Our data in mice suggest that the site of intramuscular boosting with an mRNA vaccine does not substantially impact immunity or protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Baoling Ying
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chieh-Yu Liang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pritesh Desai
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Suzanne M Scheaffer
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | - Larissa B Thackray
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
7
|
Cyster JG, Wilson PC. Antibody modulation of B cell responses-Incorporating positive and negative feedback. Immunity 2024; 57:1466-1481. [PMID: 38986442 PMCID: PMC11257158 DOI: 10.1016/j.immuni.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
Antibodies are powerful modulators of ongoing and future B cell responses. While the concept of antibody feedback has been appreciated for over a century, the topic has seen a surge in interest due to the evidence that the broadening of antibody responses to SARS-CoV-2 after a third mRNA vaccination is a consequence of antibody feedback. Moreover, the discovery that slow antigen delivery can lead to more robust humoral immunity has put a spotlight on the capacity for early antibodies to augment B cell responses. Here, we review the mechanisms whereby antibody feedback shapes B cell responses, integrating findings in humans and in mouse models. We consider the major influence of epitope masking and the diverse actions of complement and Fc receptors and provide a framework for conceptualizing the ways antigen-specific antibodies may influence B cell responses to any form of antigen, in conditions as diverse as infectious disease, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA.
| | - Patrick C Wilson
- Drukier Institute for Children's Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
8
|
Wiehe K, Saunders KO, Stalls V, Cain DW, Venkatayogi S, Martin Beem JS, Berry M, Evangelous T, Henderson R, Hora B, Xia SM, Jiang C, Newman A, Bowman C, Lu X, Bryan ME, Bal J, Sanzone A, Chen H, Eaton A, Tomai MA, Fox CB, Tam YK, Barbosa C, Bonsignori M, Muramatsu H, Alam SM, Montefiori DC, Williams WB, Pardi N, Tian M, Weissman D, Alt FW, Acharya P, Haynes BF. Mutation-guided vaccine design: A process for developing boosting immunogens for HIV broadly neutralizing antibody induction. Cell Host Microbe 2024; 32:693-709.e7. [PMID: 38670093 DOI: 10.1016/j.chom.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 01/05/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
A major goal of HIV-1 vaccine development is the induction of broadly neutralizing antibodies (bnAbs). Although success has been achieved in initiating bnAb B cell lineages, design of boosting immunogens that select for bnAb B cell receptors with improbable mutations required for bnAb affinity maturation remains difficult. Here, we demonstrate a process for designing boosting immunogens for a V3-glycan bnAb B cell lineage. The immunogens induced affinity-matured antibodies by selecting for functional improbable mutations in bnAb precursor knockin mice. Moreover, we show similar success in prime and boosting with nucleoside-modified mRNA-encoded HIV-1 envelope trimer immunogens, with improved selection by mRNA immunogens of improbable mutations required for bnAb binding to key envelope glycans. These results demonstrate the ability of both protein and mRNA prime-boost immunogens for selection of rare B cell lineage intermediates with neutralizing breadth after bnAb precursor expansion, a key proof of concept and milestone toward development of an HIV-1 vaccine.
Collapse
Affiliation(s)
- Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Department of Microbiology and Molecular Genetics, Duke University School of Medicine, Durham, NC 27710, USA; Department of Integrative Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Victoria Stalls
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Derek W Cain
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sravani Venkatayogi
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Joshua S Martin Beem
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Madison Berry
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tyler Evangelous
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Rory Henderson
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Bhavna Hora
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Shi-Mao Xia
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Chuancang Jiang
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Amanda Newman
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Cindy Bowman
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaozhi Lu
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mary E Bryan
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Joena Bal
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Aja Sanzone
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Haiyan Chen
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Amanda Eaton
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mark A Tomai
- Corporate Research Materials Lab, 3M Company, St. Paul, MN 55144, USA
| | | | | | | | - Mattia Bonsignori
- Translational Immunobiology Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hiromi Muramatsu
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - S Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - David C Montefiori
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Wilton B Williams
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Department of Integrative Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Norbert Pardi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ming Tian
- Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Frederick W Alt
- Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Priyamvada Acharya
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Department of Integrative Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
9
|
Zareein A, Mahmoudi M, Jadhav SS, Wilmore J, Wu Y. Biomaterial engineering strategies for B cell immunity modulations. Biomater Sci 2024; 12:1981-2006. [PMID: 38456305 PMCID: PMC11019864 DOI: 10.1039/d3bm01841e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
B cell immunity has a penetrating effect on human health and diseases. Therapeutics aiming to modulate B cell immunity have achieved remarkable success in combating infections, autoimmunity, and malignancies. However, current treatments still face significant limitations in generating effective long-lasting therapeutic B cell responses for many conditions. As the understanding of B cell biology has deepened in recent years, clearer regulation networks for B cell differentiation and antibody production have emerged, presenting opportunities to overcome current difficulties and realize the full therapeutic potential of B cell immunity. Biomaterial platforms have been developed to leverage these emerging concepts to augment therapeutic humoral immunity by facilitating immunogenic reagent trafficking, regulating T cell responses, and modulating the immune microenvironment. Moreover, biomaterial engineering tools have also advanced our understanding of B cell biology, further expediting the development of novel therapeutics. In this review, we will introduce the general concept of B cell immunobiology and highlight key biomaterial engineering strategies in the areas including B cell targeted antigen delivery, sustained B cell antigen delivery, antigen engineering, T cell help optimization, and B cell suppression. We will also discuss our perspective on future biomaterial engineering opportunities to leverage humoral immunity for therapeutics.
Collapse
Affiliation(s)
- Ali Zareein
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Mina Mahmoudi
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Shruti Sunil Jadhav
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
| | - Joel Wilmore
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
10
|
Syeda MZ, Hong T, Huang C, Huang W, Mu Q. B cell memory: from generation to reactivation: a multipronged defense wall against pathogens. Cell Death Discov 2024; 10:117. [PMID: 38453885 PMCID: PMC10920759 DOI: 10.1038/s41420-024-01889-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024] Open
Abstract
Development of B cell memory is a conundrum that scientists are still exploring. Studies have been conducted in vitro and using advanced animal models to elucidate the mechanism underlying the generation of memory B cells (MBCs), the precise roles of MBCs against pathogens, and their protective functions against repeated infections throughout life. Lifelong immunity against invading diseases is mainly the result of overcoming a single infection. This protection is largely mediated by the two main components of B cell memory-MBCs and long-lived plasma cells (PCs). The chemical and cellular mechanisms that encourage fat selection for MBCs or long-lived PCs are an area of active research. Despite the fact that nearly all available vaccinations rely on the capacity to elicit B-cell memory, we have yet to develop successful vaccines that can induce broad-scale protective MBCs against some of the deadliest diseases, including malaria and AIDS. A deeper understanding of the specific cellular and molecular pathways that govern the generation, function, and reactivation of MBCs is critical for overcoming the challenges associated with vaccine development. Here, we reviewed literature on the development of MBCs and their reactivation, interaction with other cell types, strategies against invading pathogens, and function throughout life and discussed the recent advances regarding the key signals and transcription factors which regulate B cell memory and their relevance to the quest for vaccine development.
Collapse
Affiliation(s)
- Madiha Zahra Syeda
- The People's Hospital of Gaozhou, Guangdong Medical University, Maoming, 525200, China
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tu Hong
- The First Affiliated Hospital, Zhejiang University, School of Medicine, 310058, Hangzhou, China
| | - Chunming Huang
- The People's Hospital of Gaozhou, Guangdong Medical University, Maoming, 525200, China.
| | - Wenhua Huang
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Qingchun Mu
- The People's Hospital of Gaozhou, Guangdong Medical University, Maoming, 525200, China.
| |
Collapse
|
11
|
Jiang W, Maldeney AR, Yuan X, Richer MJ, Renshaw SE, Luo W. Ipsilateral immunization after a prior SARS-CoV-2 mRNA vaccination elicits superior B cell responses compared to contralateral immunization. Cell Rep 2024; 43:113665. [PMID: 38194344 PMCID: PMC10851277 DOI: 10.1016/j.celrep.2023.113665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/05/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024] Open
Abstract
mRNA vaccines have proven to be pivotal in the fight against COVID-19. A recommended booster, given 3 to 4 weeks post the initial vaccination, can substantially amplify protective antibody levels. Here, we show that, compared to contralateral boost, ipsilateral boost of the SARS-CoV-2 mRNA vaccine induces more germinal center B cells (GCBCs) specific to the receptor binding domain (RBD) and generates more bone marrow plasma cells. Ipsilateral boost can more rapidly generate high-affinity RBD-specific antibodies with improved cross-reactivity to the Omicron variant. Mechanistically, the ipsilateral boost promotes the positive selection and plasma cell differentiation of pre-existing GCBCs from the prior vaccination, associated with the expansion of T follicular helper cells. Furthermore, we show that ipsilateral immunization with an unrelated antigen after a prior mRNA vaccination enhances the germinal center and antibody responses to the new antigen compared to contralateral immunization. These findings propose feasible approaches to optimize vaccine effectiveness.
Collapse
Affiliation(s)
- Wenxia Jiang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander R Maldeney
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xue Yuan
- Department of Otolaryngology - Head and Neck Surgery, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Martin J Richer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Cooperative Center of Excellence in Hematology (CCEH), Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Scott E Renshaw
- Department of Family Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Wei Luo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Cooperative Center of Excellence in Hematology (CCEH), Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
12
|
Fazli S, Thomas A, Estrada AE, Ross HA, Xthona Lee D, Kazmierczak S, Slifka MK, Montefiori D, Messer WB, Curlin ME. Contralateral second dose improves antibody responses to a 2-dose mRNA vaccination regimen. J Clin Invest 2024; 134:e176411. [PMID: 38227381 PMCID: PMC10940087 DOI: 10.1172/jci176411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/09/2024] [Indexed: 01/17/2024] Open
Abstract
BACKGROUNDVaccination is typically administered without regard to site of prior vaccination, but this factor may substantially affect downstream immune responses.METHODSWe assessed serological responses to initial COVID-19 vaccination in baseline seronegative adults who received second-dose boosters in the ipsilateral or contralateral arm relative to initial vaccination. We measured serum SARS-CoV-2 spike-specific Ig, receptor-binding domain-specific (RBD-specific) IgG, SARS-CoV-2 nucleocapsid-specific IgG, and neutralizing antibody titers against SARS-CoV-2.D614G (early strain) and SARS-CoV-2.B.1.1.529 (Omicron) at approximately 0.6, 8, and 14 months after boosting.RESULTSIn 947 individuals, contralateral boosting was associated with higher spike-specific serum Ig, and this effect increased over time, from a 1.1-fold to a 1.4-fold increase by 14 months (P < 0.001). A similar pattern was seen for RBD-specific IgG. Among 54 pairs matched for age, sex, and relevant time intervals, arm groups had similar antibody levels at study visit 2 (W2), but contralateral boosting resulted in significantly higher binding and neutralizing antibody titers at W3 and W4, with progressive increase over time, ranging from 1.3-fold (total Ig, P = 0.007) to 4.0-fold (pseudovirus neutralization to B.1.1.529, P < 0.001).CONCLUSIONSIn previously unexposed adults receiving an initial vaccine series with the BNT162b2 mRNA COVID-19 vaccine, contralateral boosting substantially increases antibody magnitude and breadth at times beyond 3 weeks after vaccination. This effect should be considered during arm selection in the context of multidose vaccine regimens.FUNDINGM.J. Murdock Charitable Trust, OHSU Foundation, NIH.
Collapse
Affiliation(s)
| | - Archana Thomas
- Oregon National Primate Research Center, Division of Neuroscience, and
| | - Abram E. Estrada
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | | | - David Xthona Lee
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Steven Kazmierczak
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, USA
| | - Mark K. Slifka
- Oregon National Primate Research Center, Division of Neuroscience, and
| | - David Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - William B. Messer
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
- Department of Medicine, Division of Infectious Diseases, Oregon Health & Science University, Portland, Oregon, USA
- Program in Epidemiology, Oregon Health & Science University, Portland State University School of Public Health, Portland, Oregon, USA
| | - Marcel E. Curlin
- Department of Occupational Health
- Department of Medicine, Division of Infectious Diseases, Oregon Health & Science University, Portland, Oregon, USA
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
13
|
Abstract
Recent advances in studies of immune memory in mice and humans have reinforced the concept that memory B cells play a critical role in protection against repeated infections, particularly from variant viruses. Hence, insights into the development of high-quality memory B cells that can generate broadly neutralizing antibodies that bind such variants are key for successful vaccine development. Here, we review the cellular and molecular mechanisms by which memory B cells are generated and how these processes shape the antibody diversity and breadth of memory B cells. Then, we discuss the mechanisms of memory B cell reactivation in the context of established immune memory; the contribution of antibody feedback to this process has now begun to be reappreciated.
Collapse
Affiliation(s)
- Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, Japan.
| |
Collapse
|
14
|
Yan M, Xiao LY, Gosau M, Friedrich RE, Smeets R, Fu LL, Feng HC, Burg S. The causal association between COVID-19 and herpes simplex virus: a Mendelian randomization study. Front Immunol 2023; 14:1281292. [PMID: 38146366 PMCID: PMC10749317 DOI: 10.3389/fimmu.2023.1281292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/28/2023] [Indexed: 12/27/2023] Open
Abstract
Introduction The coronavirus disease 2019 (COVID-19) has emerged as a main global public health challenge. Additionally, herpes simplex virus type-1 (HSV-1) and type 2 (HSV-2) are widespread viruses that can cause orolabial herpes and genital herpes. Several clinical case reports have declared a possible association between the two, however, the causal relationship between them has not been clarified. Methods This study utilized a Mendelian randomization (MR) approach for causality assessment between COVID-19 infection and HSV infection based on the latest public health data and Genome-Wide Association Study (GWAS) data. Multiple causal estimation methods, such as IVW, weighted median, simple mode, and weighted mode, were employed to validate the causal relation between COVID-19 infection and HSV infection, with COVID-19 infection, COVID-19 hospitalization, and severe COVID-19 as exposures, and HSV1/2 infection as the outcome. A reverse MR analysis was subsequently performed. Results MR analysis exhibited that COVID-19 infection was relevant to a reduced risk of HSV1 infection (p=7.603239e-152, OR=0.5690, 95%CI=0.5455-0.5935, IVW). Regarding the effect of COVID-19 infection on HSV2, MR analysis suggested that COVID-19 infection was correlated with an augmented risk of HSV2 infection (p=6.46735e-11, OR=1.1137, 95%CI=1.0782-1.1502, IVW). The reverse MR analysis did not demonstrate a reverse causal relationship between HSV and COVID-19. Discussion Altogether, COVID-19 infection might cause a decreased risk of HSV1 infection and an elevated risk of HSV2 infection.
Collapse
Affiliation(s)
- Ming Yan
- Department of Oral and Maxillofacial Surgery, Guiyang Hospital of Stomatology, Guiyang, China
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Li-yuan Xiao
- Department of Oral and Maxillofacial Surgery, Guiyang Hospital of Stomatology, Guiyang, China
| | - Martin Gosau
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Reinhard E. Friedrich
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Oral and Maxillofacial Surgery, Division of Regenerative Orofacial Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ling-ling Fu
- Department of Oral and Maxillofacial Surgery, Guiyang Hospital of Stomatology, Guiyang, China
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hong-chao Feng
- Department of Oral and Maxillofacial Surgery, Guiyang Hospital of Stomatology, Guiyang, China
| | - Simon Burg
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
15
|
Ziegler L, Klemis V, Schmidt T, Schneitler S, Baum C, Neumann J, Becker SL, Gärtner BC, Sester U, Sester M. Is ipsilateral administration of COVID-19 vaccine boosters the optimal approach? EBioMedicine 2023; 98:104853. [PMID: 38251468 PMCID: PMC10628342 DOI: 10.1016/j.ebiom.2023.104853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 01/23/2024] Open
Affiliation(s)
- Laura Ziegler
- Department of Transplant and Infection Immunology, Saarland University, Germany
| | - Verena Klemis
- Department of Transplant and Infection Immunology, Saarland University, Germany
| | - Tina Schmidt
- Department of Transplant and Infection Immunology, Saarland University, Germany
| | - Sophie Schneitler
- Department of Medical Microbiology and Hygiene, Saarland University, Germany
| | - Christina Baum
- Occupational Health Care Center, Saarland University, 66421, Homburg, Germany
| | - Jürgen Neumann
- Department of Occupational Health, Robert Bosch GmbH, 66424, Homburg, Germany
| | - Sören L Becker
- Department of Medical Microbiology and Hygiene, Saarland University, Germany
| | - Barbara C Gärtner
- Department of Medical Microbiology and Hygiene, Saarland University, Germany
| | - Urban Sester
- Department of Nephrology, SHG-Klinikum Völklingen, 66333, Völklingen, Germany
| | - Martina Sester
- Department of Transplant and Infection Immunology, Saarland University, Germany.
| |
Collapse
|
16
|
Ziegler L, Klemis V, Schmidt T, Schneitler S, Baum C, Neumann J, Becker SL, Gärtner BC, Sester U, Sester M. Differences in SARS-CoV-2 specific humoral and cellular immune responses after contralateral and ipsilateral COVID-19 vaccination. EBioMedicine 2023; 95:104743. [PMID: 37574375 PMCID: PMC10505826 DOI: 10.1016/j.ebiom.2023.104743] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/02/2023] [Accepted: 07/21/2023] [Indexed: 08/15/2023] Open
Abstract
BACKGROUND Individual doses of dual-dose vaccine-regimens are sequentially administered into the deltoid muscle, but little attention has so far been paid to the immunological effects of choosing the ipsilateral or the contralateral side for the second dose. METHODS In an observational study, 303 previously naive individuals were recruited, who received the second dose of the COVID-19 vaccine BNT162b2 on either the ipsilateral (n = 147) or the contralateral side (n = 156). Spike-specific IgG, IgG-avidity, and neutralizing antibodies were quantified using ELISA and a surrogate assay 2 weeks after dose 2. A subgroup of 143 individuals (64 ipsilateral, 79 contralateral) was analysed for spike-specific CD4 and CD8 T-cells using flow-cytometry. FINDINGS Median spike-specific IgG-levels did not differ after ipsilateral (4590 (IQR 3438) BAU/ml) or contralateral vaccination (4002 (IQR 3524) BAU/ml, p = 0.106). IgG-avidity was also similar (p = 0.056). However, neutralizing activity was significantly lower after contralateral vaccination (p = 0.024). Likewise, median spike-specific CD8 T-cell levels were significantly lower (p = 0.004). Consequently, the percentage of individuals with detectable CD8 T-cells was significantly lower after contralateral than after ipsilateral vaccination (43.0% versus 67.2%, p = 0.004). Spike specific CD4 T-cell levels were similar in both groups, but showed significantly higher CTLA-4 expression after contralateral vaccination (p = 0.011). These effects were vaccine-specific, as polyclonally stimulated T-cell levels did not differ. INTERPRETATION Both ipsilateral and contralateral vaccination induce a strong immune response, but secondary boosting is more pronounced when choosing vaccine administration-routes that allows for drainage by the same lymph nodes used for priming. Higher neutralizing antibody activity and higher levels of spike-specific CD8 T-cells may have implications for protection from infection and severe disease and support general preference for ipsilateral vaccination. FUNDING Financial support was provided in part by the State chancellery of the Saarland to M.S.
Collapse
Affiliation(s)
- Laura Ziegler
- Department of Transplant and Infection Immunology, Saarland University, Germany
| | - Verena Klemis
- Department of Transplant and Infection Immunology, Saarland University, Germany
| | - Tina Schmidt
- Department of Transplant and Infection Immunology, Saarland University, Germany
| | - Sophie Schneitler
- Department of Medical Microbiology and Hygiene, Saarland University, Germany
| | - Christina Baum
- Occupational Health Care Center, Saarland University, 66421 Homburg, Germany
| | - Jürgen Neumann
- Department of Occupational Health, Robert Bosch GmbH, 66424 Homburg, Germany
| | - Sören L Becker
- Department of Medical Microbiology and Hygiene, Saarland University, Germany
| | - Barbara C Gärtner
- Department of Medical Microbiology and Hygiene, Saarland University, Germany
| | - Urban Sester
- Department of Nephrology, SHG-Klinikum Völklingen, 66333 Völklingen, Germany
| | - Martina Sester
- Department of Transplant and Infection Immunology, Saarland University, Germany.
| |
Collapse
|
17
|
Matz HC, McIntire KM, Ellebedy AH. 'Persistent germinal center responses: slow-growing trees bear the best fruits'. Curr Opin Immunol 2023; 83:102332. [PMID: 37150126 PMCID: PMC10829534 DOI: 10.1016/j.coi.2023.102332] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 05/09/2023]
Abstract
Germinal centers (GCs) are key microanatomical sites in lymphoid organs where responding B cells mature and undergo affinity-based selection. The duration of the GC reaction has long been assumed to be relatively brief, but recent studies in humans, nonhuman primates, and mice indicate that GCs can last for weeks to months after initial antigen exposure. This review examines recent studies investigating the factors that influence GC duration, including antigen persistence, T-follicular helper cells, and mode of immunization. Potential mechanisms for how persistent GCs influence the B-cell repertoire are considered. Overall, these studies provide a blueprint for how to design better vaccines that elicit persistent GC responses.
Collapse
Affiliation(s)
- Hanover C Matz
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Katherine M McIntire
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, USA.
| |
Collapse
|
18
|
Fujisawa M, Adachi Y, Onodera T, Shiwa-Sudo N, Iwata-Yoshikawa N, Nagata N, Suzuki T, Takeoka S, Takahashi Y. High-throughput isolation of SARS-CoV-2 nucleocapsid antibodies for improved antigen detection. Biochem Biophys Res Commun 2023; 673:114-120. [PMID: 37379800 PMCID: PMC10279465 DOI: 10.1016/j.bbrc.2023.06.067] [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: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
SARS-CoV-2 nucleocapsid protein (NP) is the main target for COVID-19-diagnostic PCR and antigen rapid diagnostic tests (Ag-RDTs). Ag-RDTs are more convenient than PCR tests for point-of-care testing or self-testing to identify the SARS-CoV-2 antigen. The sensitivity and specificity of this method depends mainly on the affinity and specificity of NP-binding antibodies; therefore, antigen-antibody binding is key elements for the Ag-RDTs. Here, we applied the high-throughput antibody isolation platform that has been utilized to isolate therapeutic antibodies against rare epitopes. Two NP antibodies were identified to recognize non-overlapping epitopes with high affinity. One antibody specifically binds to SARS-CoV-2 NP, and the other rapidly and tightly binds to SARS-CoV-2 NP with cross-reactivity to SARS-CoV NP. Furthermore, these antibodies were compatible with a sandwich enzyme-linked immunosorbent assay that exhibited enhanced sensitivity for NP detection compared to the previously isolated NP antibodies. Thus, the NP antibody pair is applicable to more sensitive and specific Ag-RDTs, highlighting the utility of a high-throughput antibody isolation platform for diagnostics development.
Collapse
Affiliation(s)
- Mizuki Fujisawa
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan; Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Yu Adachi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Nozomi Shiwa-Sudo
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Naoko Iwata-Yoshikawa
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Shinji Takeoka
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan; Research Institute for Science and Engineering, Waseda University, 3-4-1, Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.
| |
Collapse
|
19
|
Lamrayah M, Phelip C, Rovera R, Coiffier C, Lazhar N, Bartolomei F, Colomb E, Verrier B, Monge C, Richard S. Poloxamers Have Vaccine-Adjuvant Properties by Increasing Dissemination of Particulate Antigen at Distant Lymph Nodes. Molecules 2023; 28:4778. [PMID: 37375333 PMCID: PMC10304813 DOI: 10.3390/molecules28124778] [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: 05/24/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Vaccine technology is still facing challenges regarding some infectious diseases, which can be addressed by innovative drug delivery systems. In particular, nanoparticle-based vaccines combined with new types of adjuvants are actively explored as a platform for improving the efficacy and durability of immune protection. Here, biodegradable nanoparticles carrying an antigenic model of HIV were formulated with two combinations of poloxamers, 188/407, presenting or not presenting gelling properties, respectively. The study aimed to determine the influence of poloxamers (as a thermosensitive hydrogel or a liquid solution) on the adaptive immune response in mice. The results showed that poloxamer-based formulations were physically stable and did not induce any toxicity using a mouse dendritic cell line. Then, whole-body biodistribution studies using a fluorescent formulation highlighted that the presence of poloxamers influenced positively the dissemination profile by dragging nanoparticles through the lymphatic system until the draining and distant lymph nodes. The strong induction of specific IgG and germinal centers in distant lymph nodes in presence of poloxamers suggested that such adjuvants are promising components in vaccine development.
Collapse
Affiliation(s)
- Myriam Lamrayah
- Laboratory of Tissue Biology and Therapeutic Engineering, Institut de Biologie et Chimie des Protéines, UMR 5305, CNRS/Claude Bernard University Lyon 1, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France; (M.L.); (C.P.); (R.R.); (C.C.); (N.L.); (F.B.); (E.C.); (B.V.); (S.R.)
- Laboratory of Virology and Genetics, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Capucine Phelip
- Laboratory of Tissue Biology and Therapeutic Engineering, Institut de Biologie et Chimie des Protéines, UMR 5305, CNRS/Claude Bernard University Lyon 1, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France; (M.L.); (C.P.); (R.R.); (C.C.); (N.L.); (F.B.); (E.C.); (B.V.); (S.R.)
| | - Renaud Rovera
- Laboratory of Tissue Biology and Therapeutic Engineering, Institut de Biologie et Chimie des Protéines, UMR 5305, CNRS/Claude Bernard University Lyon 1, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France; (M.L.); (C.P.); (R.R.); (C.C.); (N.L.); (F.B.); (E.C.); (B.V.); (S.R.)
| | - Céline Coiffier
- Laboratory of Tissue Biology and Therapeutic Engineering, Institut de Biologie et Chimie des Protéines, UMR 5305, CNRS/Claude Bernard University Lyon 1, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France; (M.L.); (C.P.); (R.R.); (C.C.); (N.L.); (F.B.); (E.C.); (B.V.); (S.R.)
| | - Nora Lazhar
- Laboratory of Tissue Biology and Therapeutic Engineering, Institut de Biologie et Chimie des Protéines, UMR 5305, CNRS/Claude Bernard University Lyon 1, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France; (M.L.); (C.P.); (R.R.); (C.C.); (N.L.); (F.B.); (E.C.); (B.V.); (S.R.)
| | - Francesca Bartolomei
- Laboratory of Tissue Biology and Therapeutic Engineering, Institut de Biologie et Chimie des Protéines, UMR 5305, CNRS/Claude Bernard University Lyon 1, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France; (M.L.); (C.P.); (R.R.); (C.C.); (N.L.); (F.B.); (E.C.); (B.V.); (S.R.)
| | - Evelyne Colomb
- Laboratory of Tissue Biology and Therapeutic Engineering, Institut de Biologie et Chimie des Protéines, UMR 5305, CNRS/Claude Bernard University Lyon 1, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France; (M.L.); (C.P.); (R.R.); (C.C.); (N.L.); (F.B.); (E.C.); (B.V.); (S.R.)
| | - Bernard Verrier
- Laboratory of Tissue Biology and Therapeutic Engineering, Institut de Biologie et Chimie des Protéines, UMR 5305, CNRS/Claude Bernard University Lyon 1, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France; (M.L.); (C.P.); (R.R.); (C.C.); (N.L.); (F.B.); (E.C.); (B.V.); (S.R.)
| | - Claire Monge
- Laboratory of Tissue Biology and Therapeutic Engineering, Institut de Biologie et Chimie des Protéines, UMR 5305, CNRS/Claude Bernard University Lyon 1, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France; (M.L.); (C.P.); (R.R.); (C.C.); (N.L.); (F.B.); (E.C.); (B.V.); (S.R.)
| | - Sophie Richard
- Laboratory of Tissue Biology and Therapeutic Engineering, Institut de Biologie et Chimie des Protéines, UMR 5305, CNRS/Claude Bernard University Lyon 1, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France; (M.L.); (C.P.); (R.R.); (C.C.); (N.L.); (F.B.); (E.C.); (B.V.); (S.R.)
| |
Collapse
|
20
|
Reusch L, Angeletti D. Memory B-cell diversity: From early generation to tissue residency and reactivation. Eur J Immunol 2023; 53:e2250085. [PMID: 36811174 DOI: 10.1002/eji.202250085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/17/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023]
Abstract
Memory B cells (MBCs) have a crucial function in providing an enhanced response to repeated infections. Upon antigen encounter, MBC can either rapidly differentiate to antibody secreting cells or enter germinal centers (GC) to further diversify and affinity mature. Understanding how and when MBC are formed, where they reside and how they select their fate upon reactivation has profound implications for designing strategies to improve targeted, next-generation vaccines. Recent studies have crystallized much of our knowledge on MBC but also reported several surprising discoveries and gaps in our current understanding. Here, we review the latest advancements in the field and highlight current unknowns. In particular, we focus on timing and cues leading to MBC generation before and during the GC reaction, discuss how MBC become resident in mucosal tissues, and finally, provide an overview of factors shaping MBC fate-decision upon reactivation in mucosal and lymphoid tissues.
Collapse
Affiliation(s)
- Laura Reusch
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Davide Angeletti
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
21
|
Schiepers A, van 't Wout MFL, Greaney AJ, Zang T, Muramatsu H, Lin PJC, Tam YK, Mesin L, Starr TN, Bieniasz PD, Pardi N, Bloom JD, Victora GD. Molecular fate-mapping of serum antibody responses to repeat immunization. Nature 2023; 615:482-489. [PMID: 36646114 PMCID: PMC10023323 DOI: 10.1038/s41586-023-05715-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023]
Abstract
The protective efficacy of serum antibodies results from the interplay of antigen-specific B cell clones of different affinities and specificities. These cellular dynamics underlie serum-level phenomena such as original antigenic sin (OAS)-a proposed propensity of the immune system to rely repeatedly on the first cohort of B cells engaged by an antigenic stimulus when encountering related antigens, in detriment to the induction of de novo responses1-5. OAS-type suppression of new, variant-specific antibodies may pose a barrier to vaccination against rapidly evolving viruses such as influenza and SARS-CoV-26,7. Precise measurement of OAS-type suppression is challenging because cellular and temporal origins cannot readily be ascribed to antibodies in circulation; its effect on subsequent antibody responses therefore remains unclear5,8. Here we introduce a molecular fate-mapping approach with which serum antibodies derived from specific cohorts of B cells can be differentially detected. We show that serum responses to sequential homologous boosting derive overwhelmingly from primary cohort B cells, while later induction of new antibody responses from naive B cells is strongly suppressed. Such 'primary addiction' decreases sharply as a function of antigenic distance, allowing reimmunization with divergent viral glycoproteins to produce de novo antibody responses targeting epitopes that are absent from the priming variant. Our findings have implications for the understanding of OAS and for the design and testing of vaccines against evolving pathogens.
Collapse
Affiliation(s)
- Ariën Schiepers
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | | | - Allison J Greaney
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Trinity Zang
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paulo J C Lin
- Acuitas Therapeutics, Vancouver, British Columbia, Canada
| | - Ying K Tam
- Acuitas Therapeutics, Vancouver, British Columbia, Canada
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Tyler N Starr
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Paul D Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jesse D Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
22
|
Foster WS, Lee JL, Thakur N, Newman J, Spencer AJ, Davies S, Woods D, Godfrey L, Hay IM, Innocentin S, Yam-Puc JC, Horner EC, Sharpe HJ, Thaventhiran JE, Bailey D, Lambe T, Linterman MA. Tfh cells and the germinal center are required for memory B cell formation & humoral immunity after ChAdOx1 nCoV-19 vaccination. Cell Rep Med 2022; 3:100845. [PMID: 36455555 PMCID: PMC9663747 DOI: 10.1016/j.xcrm.2022.100845] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/19/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Emergence from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has been facilitated by the rollout of effective vaccines. Successful vaccines generate high-affinity plasma blasts and long-lived protective memory B cells. Here, we show a requirement for T follicular helper (Tfh) cells and the germinal center reaction for optimal serum antibody and memory B cell formation after ChAdOx1 nCoV-19 vaccination. We found that Tfh cells play an important role in expanding antigen-specific B cells while identifying Tfh-cell-dependent and -independent memory B cell subsets. Upon secondary vaccination, germinal center B cells generated during primary immunizations can be recalled as germinal center B cells again. Likewise, primary immunization GC-Tfh cells can be recalled as either Tfh or Th1 cells, highlighting the pluripotent nature of Tfh cell memory. This study demonstrates that ChAdOx1 nCoV-19-induced germinal centers are a critical source of humoral immunity.
Collapse
Affiliation(s)
- William S Foster
- Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Jia Le Lee
- Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Nazia Thakur
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK; Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford and Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), Oxford OX3 7BN, UK
| | - Joseph Newman
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK
| | - Alexandra J Spencer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Sophie Davies
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford and Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), Oxford OX3 7BN, UK
| | - Danielle Woods
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Leila Godfrey
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Iain M Hay
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | - Silvia Innocentin
- Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Juan Carlos Yam-Puc
- MRC Toxicology Unit, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Emily C Horner
- MRC Toxicology Unit, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Hayley J Sharpe
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | | - Dalan Bailey
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford and Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), Oxford OX3 7BN, UK.
| | - Michelle A Linterman
- Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
23
|
Künzli M, O’Flanagan SD, LaRue M, Talukder P, Dileepan T, Stolley JM, Soerens AG, Quarnstrom CF, Wijeyesinghe S, Ye Y, McPartlan JS, Mitchell JS, Mandl CW, Vile R, Jenkins MK, Ahmed R, Vezys V, Chahal JS, Masopust D. Route of self-amplifying mRNA vaccination modulates the establishment of pulmonary resident memory CD8 and CD4 T cells. Sci Immunol 2022; 7:eadd3075. [PMID: 36459542 PMCID: PMC9832918 DOI: 10.1126/sciimmunol.add3075] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Respiratory tract resident memory T cells (TRM), typically generated by local vaccination or infection, can accelerate control of pulmonary infections that evade neutralizing antibody. It is unknown whether mRNA vaccination establishes respiratory TRM. We generated a self-amplifying mRNA vaccine encoding the influenza A virus nucleoprotein that is encapsulated in modified dendron-based nanoparticles. Here, we report how routes of immunization in mice, including contralateral versus ipsilateral intramuscular boosts, or intravenous and intranasal routes, influenced influenza-specific cell-mediated and humoral immunity. Parabiotic surgeries revealed that intramuscular immunization was sufficient to establish CD8 TRM in the lung and draining lymph nodes. Contralateral, compared with ipsilateral, intramuscular boosting broadened the distribution of lymph node TRM and T follicular helper cells but slightly diminished resulting levels of serum antibody. Intranasal mRNA delivery established modest circulating CD8 and CD4 T cell memory but augmented distribution to the respiratory mucosa. Combining intramuscular immunizations with an intranasal mRNA boost achieved high levels of both circulating T cell memory and lung TRM. Thus, routes of mRNA vaccination influence humoral and cell-mediated immunity, and intramuscular prime-boosting establishes lung TRM that can be further expanded by an additional intranasal immunization.
Collapse
Affiliation(s)
- Marco Künzli
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stephen D. O’Flanagan
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Madeleine LaRue
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Thamotharampillai Dileepan
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - J. Michael Stolley
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew G. Soerens
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Clare F. Quarnstrom
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sathi Wijeyesinghe
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yanqi Ye
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Jason S. Mitchell
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Richard Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Marc K. Jenkins
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Vaiva Vezys
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - David Masopust
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
24
|
Haase P, Schäfer S, Gerlach RG, Winkler TH, Voehringer D. B cell fate mapping reveals their contribution to the memory immune response against helminths. Front Immunol 2022; 13:1016142. [PMID: 36505408 PMCID: PMC9730276 DOI: 10.3389/fimmu.2022.1016142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
An estimated quarter of the human world population is infected with gastrointestinal helminths causing major socioeconomic problems in endemic countries. A better understanding of humoral immune responses against helminths is urgently needed to develop effective vaccination strategies. Here, we used a fate mapping (FM) approach to mark germinal center (GC) B cells and their developmental fates by induced expression of a fluorescent protein during infection of mice with the helminth Nippostrongylus brasiliensis. We could show that FM+ cells persist weeks after clearance of the primary infection mainly as CD80+CD73+PD-L2+ memory B cells. A secondary infection elicited expansion of helminth-specific memory B cells and plasma cells (PCs). Adoptive transfers and analysis of somatic mutations in immunoglobulin genes further revealed that FM+ B cells rapidly convert to PCs rather than participating again in a GC reaction. These results provide new insights in the population dynamics of the humoral immune response against helminths.
Collapse
Affiliation(s)
- Paul Haase
- Department of Infection Biology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Simon Schäfer
- Department of Genetics, Faculty of Sciences, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Roman G. Gerlach
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Thomas H. Winkler
- Department of Genetics, Faculty of Sciences, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, Universitätsklinikum Erlangen, Erlangen, Germany,Faculty of Medicine, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany,*Correspondence: David Voehringer,
| |
Collapse
|
25
|
Valeri V, Sochon A, Ye C, Mao X, Lecoeuche D, Fillatreau S, Weill JC, Reynaud CA, Hao Y. B cell intrinsic and extrinsic factors impacting memory recall responses to SRBC challenge. Front Immunol 2022; 13:873886. [PMID: 35967317 PMCID: PMC9367638 DOI: 10.3389/fimmu.2022.873886] [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: 02/11/2022] [Accepted: 07/06/2022] [Indexed: 11/20/2022] Open
Abstract
MBCs (MBCs) generated in T-dependent immune responses can persist for a lifetime and rapidly react upon secondary antigen exposure to differentiate into plasma cells (PCs) and/or to improve the affinity of their BCR through new rounds of hypermutation in germinal centers (GCs). The fate of a MBC in secondary immune reactions appears to depend upon multiple parameters, whose understanding is mandatory for the design of efficient vaccine strategies. We followed the behavior of MBCs in recall responses to SRBCs using an inducible AID fate mapping mouse model in which B cells engaged in a germinal center (GC) response are irreversibly labeled upon simultaneous tamoxifen ingestion and immunization. We used different schemes of mouse immunization and tamoxifen feeding in adoptive-transfer experiments of total splenic B cells into congenic mice that have been pre-immunized or not, to assess the contribution of the different effector subsets in a physiological competitive context. We were able to show that naive B cells can differentiate into GC B cells with kinetics similar to MBCs in the presence of previously activated T follicular helper (TFH) cells and a primed microenvironment. We also showed that MBCs are recruited into secondary GCs, together with naive B cells. In contrast, PC differentiation, which dominated secondary MBC responses, was not dependent upon a previous TFH activation. We observed that the presence of persisting germinal centers and circulating antibody levels are key factors determining the germinal center versus plasma cell fate in a recall response. Notably, disruption of persistent germinal center structures by a lymphotoxin beta-receptor fusion protein or a longer timing between the prime and the boost, which correlated with reduced antigen-specific immunoglobulin levels in serum, were two conditions with an opposite impact, respectively inhibiting or promoting a GC fate for MBCs. Altogether, these studies highlight the complexity of recall responses, whose outcome varies according to immunization contexts.
Collapse
Affiliation(s)
- Viviana Valeri
- Institut Necker Enfants-Malades, INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Akhésa Sochon
- Institut Necker Enfants-Malades, INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Chaoliang Ye
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinru Mao
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Damiana Lecoeuche
- Institut Necker Enfants-Malades, INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Simon Fillatreau
- Institut Necker Enfants-Malades, INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Jean-Claude Weill
- Institut Necker Enfants-Malades, INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Claude-Agnès Reynaud
- Institut Necker Enfants-Malades, INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Yi Hao
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|