1
|
Harmsen MM, Harders F, van de Water S, van Solt CS, Koets AP, van der Wal FJ, Gupta N. Optimization of blood sampling time for isolation of antigen binding single-domain antibodies from immunized llamas. J Immunol Methods 2025; 539:113844. [PMID: 40044074 DOI: 10.1016/j.jim.2025.113844] [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: 11/11/2024] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 03/09/2025]
Abstract
Single-domain antibody fragments (VHHs) binding specific antigens are often isolated by llama immunization and phage display selection. We determined the optimal blood sampling time for generating phage display libraries with a high frequency of antigen-binding VHHs. Two llamas were immunized with 7 antigens. Blood samples collected with 2-3 day interval up to 11 days post immunization were used for deep sequencing and phage display selection of VHHs. The frequency of VHH clones was determined by the identification of unique VHH sequences in deep sequenced libraries. On average, the highest clone frequency was found in libraries generated 5 days post immunization. Polyclonal phage ELISA also suggested day 5 as the optimal blood sampling time.
Collapse
Affiliation(s)
- Michiel M Harmsen
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, the Netherlands
| | - Frank Harders
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, the Netherlands
| | - Sandra van de Water
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, the Netherlands
| | - Conny S van Solt
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, the Netherlands
| | - Ad P Koets
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, the Netherlands
| | - Fimme J van der Wal
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, the Netherlands.
| | - Nishi Gupta
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, the Netherlands
| |
Collapse
|
2
|
Castañeda J, Poblete L, Rosemblatt MV, Sauma D, Rosemblatt M, Bono MR, Nuñez S. Early-life homeostatic differentiation of thymus-resident B cells into memory B cells. Front Immunol 2025; 16:1567788. [PMID: 40226628 PMCID: PMC11985456 DOI: 10.3389/fimmu.2025.1567788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
The thymus contains various antigen-presenting cells, including B cells, which remain activated even under steady-state conditions, suggesting ongoing local stimulation. In this study, we identify class-switched memory B cells in the thymus. Some of these cells switch their immunoglobulin to IgG2b and IgA, and express typical memory markers CD73 and PD-L2. Memory B cell differentiation in the thymus begins in neonatal mice, preceding the appearance of class-switched B cells in other peripheral lymphoid organs. Notably, exposure to environmental antigens does not influence their differentiation. Additionally, cognate interaction with CD4+ positive thymocytes is crucial for the development of memory B cells in the thymus. Our findings demonstrate that the thymus supports the local differentiation of memory B cells through a steady-state process, independent of foreign antigen stimulation and driven by interactions with developing T cells.
Collapse
Affiliation(s)
- Justine Castañeda
- Escuela de Postgrado, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Lilian Poblete
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Mariana V. Rosemblatt
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Los Leones, Santiago, Chile
| | - Daniela Sauma
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| | - Mario Rosemblatt
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Los Leones, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| | - María Rosa Bono
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| | - Sarah Nuñez
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Los Leones, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| |
Collapse
|
3
|
Lu B, Chaudhary O, Banoth B, Nadkarni J, Zong W, Mausser E, Danz H, Motwani M, Ruiz S, Zhang D, Nageshwaran G, Rokbi B, Warren W, DeRosa F, Chivukula S. Impact of Extended Dosing Intervals and Ipsilateral Versus Contralateral Boosting on mRNA Vaccine Immunogenicity in Mice. Vaccines (Basel) 2025; 13:263. [PMID: 40266125 PMCID: PMC11946721 DOI: 10.3390/vaccines13030263] [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: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Although mRNA vaccines have the potential to be developed and deployed rapidly to combat infectious diseases, the ideal method of administration and boosting schedule strategy for generating optimal immunogenicity is an area of active research. We compared the immune responses resulting from different schedules for prime-boost and boosting either ipsilaterally or contralaterally in relation to the initial vaccine dose. METHODS Influenza hemagglutinin (HA) was used as a model antigen for different vaccination regimens in mice using both mRNA lipid nanoparticles (mRNA-LNP) and AF03-adjuvanted recombinant protein (rHA-AF03) vaccines. RESULTS Increasing the prime-boost interval resulted in higher levels of serum anti-HA IgG and functional antibody hemagglutination inhibition (HAI) responses in mRNA-LNP-vaccinated animals, which correlated with an induction of germinal center (GC) B cells and follicular helper T (Tfh) cells in lymph nodes. In addition, longer prime-boost intervals resulted in higher levels of IL-2 and TNF-α producing CD4+ T cells two weeks after boosting. The number of Ig-secreting long-lived plasma cells increased with the length of prime-boost intervals. Contralateral boosting resulted in an increase in HAI titers and GC B cells compared to an ipsilateral boost. However, significantly higher numbers of GC B cells were induced in the draining lymph nodes following ipsilateral boosting than in the non-draining lymph nodes. CONCLUSIONS Overall, our data provides insights into the immune mechanisms of action of mRNA-LNP to develop the optimal vaccine regimen for mRNA vaccine platforms.
Collapse
Affiliation(s)
- Bin Lu
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Omkar Chaudhary
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Balaji Banoth
- Former Employee of Sanofi, 200 West St., Waltham, MA 02451, USA
| | - Janhavi Nadkarni
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Wei Zong
- Translational and Early Development Biostatistics, Sanofi, 200 West St., Waltham, MA 02451, USA (D.Z.)
| | - Emilie Mausser
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Hillary Danz
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Mona Motwani
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Sophie Ruiz
- mRNA Center of Excellence, Sanofi, 1541 Avenue Marcel Mérieux, 69280 MarcyL’Etoile, France;
| | - Donghui Zhang
- Translational and Early Development Biostatistics, Sanofi, 200 West St., Waltham, MA 02451, USA (D.Z.)
| | - Gopinath Nageshwaran
- Global Antigen Design, Sanofi, 200 West St., Waltham, MA 02451, USA; (G.N.); (W.W.)
| | - Bachra Rokbi
- Global Antigen Design, Sanofi, 1541 Avenue Marcel Mérieux, 69280 Marcy L’Etoile, France;
| | - William Warren
- Global Antigen Design, Sanofi, 200 West St., Waltham, MA 02451, USA; (G.N.); (W.W.)
| | - Frank DeRosa
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Sudha Chivukula
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| |
Collapse
|
4
|
Gol Mohammad Pour Afrakoti L, Daneshpour Moghadam S, Hadinezhad P. Alzheimer's disease and the immune system: A comprehensive overview with a focus on B cells, humoral immunity, and immunotherapy. J Alzheimers Dis Rep 2025; 9:25424823251329188. [PMID: 40297057 PMCID: PMC12035277 DOI: 10.1177/25424823251329188] [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: 05/08/2024] [Accepted: 02/11/2025] [Indexed: 04/30/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder and the major cause of dementia. Amyloid-β (Aβ) and tau aggregation, mitochondrial dysfunction, and microglial dysregulation are key contributors to AD pathogenesis. Impairments in the blood-brain barrier have unveiled the contribution of the immune system, particularly B cells, in AD pathology. B cells, a crucial component of adaptive immunity, exhibit diverse functions, including antigen presentation and antibody production. While their role in neuroinflammatory disorders has been well-documented, their specific function in AD lacks adequate data. This review examines the dual role of the B cells and humoral immunity in modulating brain inflammation in AD and explores recent advancements in passive and active immunotherapeutic strategies targeting AD pathobiology. We summarize preclinical and clinical studies investigating B cell frequency, altered antibody levels, and their implications in neuroinflammation and immunotherapy. Notably, B cells demonstrate protective and pathological roles in AD, influencing neurodegeneration through antibody-mediated clearance of toxic aggregates and inflammatory activation inflammation. Passive immunotherapies targeting Aβ have shown potential in reducing amyloid plaques, while active immunotherapies are emerging as promising strategies, requiring further validation. Understanding the interplay between B cells, humoral immunity, microglia, and mitochondrial dysfunction is critical to unraveling AD pathogenesis. Their dual nature in disease progression underscores the need for precise therapeutic interventions to optimize immunotherapy outcomes and mitigate neuroinflammation effectively.
Collapse
Affiliation(s)
| | - Sanam Daneshpour Moghadam
- Department of Diagnostic and Public Health, School of Biotechnology, University of Verona, Verona, Italy
| | - Pezhman Hadinezhad
- Cognitive Neurology, Dementia and Neuropsychiatry Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Pinto TNC, Benard G, Fernandes JR. Phenotypic Characterization of B-Lymphocyte Subpopulations in Human Peripheral Blood: A Cost-Effective Seven-Color One-Tube Protocol. Methods Mol Biol 2025; 2857:15-31. [PMID: 39348052 DOI: 10.1007/978-1-0716-4128-6_3] [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: 10/01/2024]
Abstract
B cells are crucial components of the immune system, responsible for producing specific antibodies in response to infections and vaccines. Despite their uniform appearance, B cells display diverse surface molecules and functional properties, which are not yet fully understood. Apart from antibody production, B cells also play roles in antigen presentation and cytokine secretion, essential for initiating T-cell immune responses. Their significance as disease biomarkers and therapeutic targets has led to increased research focus. However, the lack of standardized protocols for B-cell identification and the variability in defining B-lymphocyte subpopulations pose some challenges. This paper proposes a B-cell identification panel throughout the evaluation of previous cytometry panels and nomenclature heterogeneity for B-cell subpopulations. Major subpopulations recognized in human peripheral blood include transitional, naive, switched memory, unswitched memory, double negative, and plasmablasts, characterized based on their functional and phenotypic features. We present a standardized flow cytometry protocol utilizing surface phenotypic markers (CD3, CD19, IgD, CD27, CD38, and CD24) to differentiate and analyze B-cell subpopulations. This practical and cost-effective panel can be used in various research and laboratory settings. The challenges of standardizing names and markers for classifying B-lymphocyte subpopulations are discussed, along with protocols utilizing multiple markers and gating strategies, allied with the importance of considering viability markers. In summary, this standardized protocol and panel provide a comprehensive approach to identifying B-cell subpopulations to enhance the reproducibility and comparability of B-cell subpopulation studies.
Collapse
Affiliation(s)
- Thalyta Nery Carvalho Pinto
- Laboratory of Dermatology and Immunodeficiencies (LIM56), Tropical Medicine Institute (IMT), School of Medicine, São Paulo University, São Paulo, Brazil
| | - Gil Benard
- Laboratory of Dermatology and Immunodeficiencies (LIM56), Tropical Medicine Institute (IMT), School of Medicine, São Paulo University, São Paulo, Brazil
| | - Juliana Ruiz Fernandes
- Laboratory of Dermatology and Immunodeficiencies (LIM56), Tropical Medicine Institute (IMT), School of Medicine, São Paulo University, São Paulo, Brazil.
| |
Collapse
|
6
|
Thakur B, Alam J, Cronin K, Patel P, Anasti K, Kane AP, Hossain A, Do H, Mansouri K, Spence TN, Edwards RJ, Janowska K, Lella M, Cook A, Saunders K, Gnanakaran S, Haynes BF, Acharya P, Alam SM. Anti-HIV-1 B cell antigen receptor signaling and structure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623645. [PMID: 39605516 PMCID: PMC11601489 DOI: 10.1101/2024.11.15.623645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The B cell antigen receptor (BCR) complex, comprised of antigen recognition and signaling components, functions in initiating B cell activation. While structural studies have described BCR domain organization, gaps remain in our understanding of its antigen binding domain (Fab, fragment antigen-binding) disposition, and how antigen binding is sensed to initiate signaling. Here, we report antigen affinity and signaling of the immunoglobulin (Ig) class IgM and IgG BCRs and define conformational states of full-length BCRs of two human broadly neutralizing antibodies, the glycan-specific, heavy chain domain-swapped, I-shaped 2G12, and a canonical Y-shaped antibody, CH31, that recognizes the CD4-binding site on the HIV-1 Envelope protein (Env). The BCRs adopted the shapes (I or Y) of their respective soluble antibodies, and both Ig class of BCRs of the same specificity bound Env trimers with similar affinities. We observed antigen-valenc y dependent differential signaling by the 2G12 IgM and IgG BCRs with trimeric Envs. Cryo-electron microscopy of the 2G12 IgG and CH31 IgM BCRs revealed varied Fab orientations. Structural comparisons revealed hinge points and regions of flexibility in the BCRs suggesting a highly dynamic structure of the BCR complex. Taken together, our results provide an integrated understanding of BCR structure, conformation, antigen recognition and signaling, and provide the basis for understanding antigen induced BCR signal transmission. One Sentence Summary Cryo-electron microscopy structures of full-length B cell antigen receptor complex provide a novel dynamic BCR model and a basis for understanding antigen binding induced signal transmission.
Collapse
|
7
|
Martin MU, Tay CM, Siew TW. Continuous Treatment with IncobotulinumtoxinA Despite Presence of BoNT/A Neutralizing Antibodies: Immunological Hypothesis and a Case Report. Toxins (Basel) 2024; 16:422. [PMID: 39453199 PMCID: PMC11510976 DOI: 10.3390/toxins16100422] [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: 06/24/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
Botulinum Neurotoxin A (BoNT/A) is a bacterial protein that has proven to be a valuable pharmaceutical in therapeutic indications and aesthetic medicine. One major concern is the formation of neutralizing antibodies (nAbs) to the core BoNT/A protein. These can interfere with the therapy, resulting in partial or complete antibody (Ab)-mediated secondary non-response (SNR) or immunoresistance. If titers of nAbs reach a level high enough that all injected BoNT/A molecules are neutralized, immunoresistance occurs. Studies have shown that continuation of treatment of neurology patients who had developed Ab-mediated partial SNR against complexing protein-containing (CPC-) BoNT/A was in some cases successful if patients were switched to complexing protein-free (CPF-) incobotulinumtoxinA (INCO). This seems to contradict the layperson's basic immunological understanding that repeated injection with the same antigen BoNT/A should lead to an increase in antigen-specific antibody titers. As such, we strive to explain how immunological memory works in general, and based on this, we propose a working hypothesis for this paradoxical phenomenon observed in some, but not all, neurology patients with immunoresistance. A critical factor is the presence of potentially immune-stimulatory components in CPC-BoNT/A products that can act as immunologic adjuvants and activate not only naïve, but also memory B lymphocyte responses. Furthermore, we propose that continuous injection of a BoN/TA formulation with low immunogenicity, e.g., INCO, may be a viable option for aesthetic patients with existing nAbs. These concepts are supported by a real-world case example of a patient with immunoresistance whose nAb levels declined with corresponding resumption of clinical response despite regular INCO injections.
Collapse
Affiliation(s)
| | | | - Tuck Wah Siew
- Radium Medical Aesthetics, 3 Temasek Boulevard #03-325/326/327/328, Suntec City Mall, Singapore 038983, Singapore
| |
Collapse
|
8
|
Lee DH, Lee J, Ahn SY, Ho TL, Kim K, Ko EJ. Monophosphoryl lipid A and poly I:C combination enhances immune responses of equine influenza virus vaccine. Vet Immunol Immunopathol 2024; 271:110743. [PMID: 38522410 DOI: 10.1016/j.vetimm.2024.110743] [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: 11/17/2023] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Abstract
Equine influenza is a contagious respiratory disease caused by H3N8 type A influenza virus. Vaccination against equine influenza is conducted regularly; however, infection still occurs globally because of the short immunity duration and suboptimal efficacy of current vaccines. Hence the objective of this study was to investigate whether an adjuvant combination can improve immune responses to equine influenza virus (EIV) vaccines. Seventy-two mice were immunized with an EIV vaccine only or with monophosphoryl lipid A (MPL), polyinosinic-polycytidylic acid (Poly I:C), or MPL + Poly I:C. Prime immunization was followed by boost immunization after 2 weeks. Mice were euthanized at 4, 8, and 32 weeks post-prime immunization, respectively. Sera were collected to determine humoral response. Bone marrow, spleen, and lung samples were harvested to determine memory cell responses, antigen-specific T-cell proliferation, and lung viral titers. MPL + Poly I:C resulted in the highest IgG, IgG1, and IgG2a antibodies and hemagglutination inhibition titers among the groups and sustained their levels until 32 weeks post-prime immunization. The combination enhanced memory B cell responses in the bone marrow and spleen. At 8 weeks post-prime immunization, the combination induced higher CD8+ central memory T cell frequencies in the lungs and CD8+ central memory T cells in the spleen. In addition, the combination group exhibited enhanced antigen-specific T cell proliferation, except for CD4+ T cells in the lungs. Our results demonstrated improved immune responses when using MPL + Poly I:C in EIV vaccines by inducing enhanced humoral responses, memory cell responses, and antigen-specific T cell proliferation.
Collapse
Affiliation(s)
- Dong-Ha Lee
- Department of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea; Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Republic of Korea
| | - Jueun Lee
- Department of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - So Yeon Ahn
- Department of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea; Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Republic of Korea
| | - Thi Len Ho
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Kiyeon Kim
- Department of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Eun-Ju Ko
- Department of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea; Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Republic of Korea; Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
9
|
Liu Y, Ren S, Ma L, Lin X, Lu J, Cao Z, Zheng S, Hu Z, Xu X, Chen X. Peg-IFNα combined with hepatitis B vaccination contributes to HBsAg seroconversion and improved immune function. Virol J 2024; 21:77. [PMID: 38555445 PMCID: PMC10981809 DOI: 10.1186/s12985-024-02344-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/17/2024] [Indexed: 04/02/2024] Open
Abstract
PURPOSE The purpose of this study was to investigate immunological variations between a group that received the hepatitis B vaccine and a non-vaccine group. We focused on a cohort that achieved HBsAg seroclearance after Peg-IFNα treatment of CHB. METHODS We enrolled twenty-eight individuals who achieved HBsAg seroclearance after Peg-IFNα treatment. They were divided into two groups: a vaccine group (n = 14) and a non-vaccine group (n = 14). We assessed lymphocyte subpopulations, B cell- and T cell-surface costimulatory/inhibitory factors, cytokines and immunoglobulin levels were detected at different time points to explore immune-function differences between both groups. RESULTS The seroconversion rate in the vaccine group at 24 weeks post-vaccination was 100%, which was significantly higher (p = 0.006) than that of the non-vaccine group (50%). Additionally, more individuals in the vaccine group exhibited anti-HBs levels exceeding 100 IUs/L and 300 IUs/L compared to the non-vaccine group (p < 0.05). The vaccine group demonstrated significantly increase total B cells and class-switched B cells at 24 weeks and plasma cells, CD80+B cells, Tfh cells, and ICOS+Tfh cell at 12 weeks, compared with baseline levels (p < 0.05). Conversely, Bregs (CD24+CD27+ and CD24+CD38high) decreased significantly at 24 weeks (p < 0.05). None of the above changes were statistically significance in the non-vaccine group (p > 0.05). Total IgG increased significantly in the vaccine group, and IL-2, IL-5, and IL-6 concentrations increased significantly at week 24 (p < 0.05). Differences in various types of cytokines and immunoglobulins in the plasma of the non-vaccine group were not significant (p > 0.05). Anti-HBs titers positively correlated with Th1/Th2 cells at 24 weeks (r = 0.448 and 0.458, respectively, p = 0.022 and 0.019, respectively), and negatively with CD24+CD38highBreg cells (r = -0.402, p = 0.042). CONCLUSIONS After achieving HBsAg seroclearance through Peg-IFNα treatment for CHB, administering the hepatitis B vaccine significantly increased anti-HBs-seroconversion rates and antibody levels. We also observed significant immunological differences between the vaccine and non-vaccine groups. Specifically, the vaccine group exhibited significant increases in B cells, plasma cells, and Tfh cells, while Breg levels was significantly lower. These immunological changes are likely conducive to the production of anti-HBs antibodies. However, in the non-vaccine group, the observed changes were not significantlly significant.
Collapse
Affiliation(s)
- Yisi Liu
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Shan Ren
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Lina Ma
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Xiao Lin
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Junfeng Lu
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Zhenhuan Cao
- Third Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Sujun Zheng
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Zhongjie Hu
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Xiaoxue Xu
- Department of Core Facility Center, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Xinyue Chen
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China.
| |
Collapse
|
10
|
Dennis E, Murach M, Blackburn CM, Marshall M, Root K, Pattarabanjird T, Deroissart J, Erickson LD, Binder CJ, Bekiranov S, McNamara CA. Loss of TET2 increases B-1 cell number and IgM production while limiting CDR3 diversity. Front Immunol 2024; 15:1380641. [PMID: 38601144 PMCID: PMC11004297 DOI: 10.3389/fimmu.2024.1380641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024] Open
Abstract
Recent studies have demonstrated a role for Ten-Eleven Translocation-2 (TET2), an epigenetic modulator, in regulating germinal center formation and plasma cell differentiation in B-2 cells, yet the role of TET2 in regulating B-1 cells is largely unknown. Here, B-1 cell subset numbers, IgM production, and gene expression were analyzed in mice with global knockout of TET2 compared to wildtype (WT) controls. Results revealed that TET2-KO mice had elevated numbers of B-1a and B-1b cells in their primary niche, the peritoneal cavity, as well as in the bone marrow (B-1a) and spleen (B-1b). Consistent with this finding, circulating IgM, but not IgG, was elevated in TET2-KO mice compared to WT. Analysis of bulk RNASeq of sort purified peritoneal B-1a and B-1b cells revealed reduced expression of heavy and light chain immunoglobulin genes, predominantly in B-1a cells from TET2-KO mice compared to WT controls. As expected, the expression of IgM transcripts was the most abundant isotype in B-1 cells. Yet, only in B-1a cells there was a significant increase in the proportion of IgM transcripts in TET2-KO mice compared to WT. Analysis of the CDR3 of the BCR revealed an increased abundance of replicated CDR3 sequences in B-1 cells from TET2-KO mice, which was more clearly pronounced in B-1a compared to B-1b cells. V-D-J usage and circos plot analysis of V-J combinations showed enhanced usage of VH11 and VH12 pairings. Taken together, our study is the first to demonstrate that global loss of TET2 increases B-1 cell number and IgM production and reduces CDR3 diversity, which could impact many biological processes and disease states that are regulated by IgM.
Collapse
Affiliation(s)
- Emily Dennis
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Maria Murach
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, United States
| | - Cassidy M.R. Blackburn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Melissa Marshall
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Katherine Root
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Tanyaporn Pattarabanjird
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Justine Deroissart
- Department for Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Loren D. Erickson
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Christoph J. Binder
- Department for Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Stefan Bekiranov
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, United States
| | - Coleen A. McNamara
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
11
|
Zhao L, Zhang Y, Guo X, Li Z, Lu W, Pan Z, Guo Y, Sun J, Zhang Y, Hao J, Guo Z. Peripheral Blood Lymphocyte Subsets in Factor VIII Inhibitor-Positive Patients with Severe Hemophilia A: A Case-Control Study. Clin Appl Thromb Hemost 2024; 30:10760296241268421. [PMID: 39149981 PMCID: PMC11329969 DOI: 10.1177/10760296241268421] [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: 05/07/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024] Open
Abstract
INTRODUCTION AND OBJECTIVES The present study aimed to investigate different peripheral lymphocyte subsets in patients with severe hemophilia A (HA) and factor VIII (FVIII) inhibitor production. For this, age-matched cases of 19 FVIII inhibitor-positive (IP), 21 FVIII inhibitor-negative (IN) and 45 healthy controls were selected for study. METHODS Flow cytometry was used to analyze the peripheral lymphocyte subsets, including T, B, natural killer (NK) and NKT cells. The T cell subsets included CD3 + CD4-CD8- [double negative T (DNT)], CD3 + CD4 + CD8+ [double-positive T (DPT)], CD3 + CD4 + CD8- and CD3 + CD4-CD8+ T cells. Pairwise comparisons of absolute lymphocyte subset values were conducted among the three groups. The cut-off value for absolute lymphocyte counts was determined using receiver operating characteristic curve analysis. RESULTS The results demonstrated that the absolute values of DPT cells in the IN and IP groups were significantly lower than those in the healthy control group (P = 0.007). The DNT values were also lower in severe HA patients with or without inhibitor than those in healthy subjects, but these differences were not statistically significant (P = 0.053). In addition, the absolute value of CD4+ Th cells in the IP group was lower than that in the healthy controls (P = 0.013). Although not statistically significant (P = 0.064), the absolute values of NKT cells were higher in the IN group compared with the IP group, and higher in the IP group compared with the healthy control group. There were no statistically significant differences in total T, B, CD8 + and NK cells among the IN, IP and healthy control groups. The cut-off value for absolute CD4+ Th cells in the IN group was < 598/µl. CONCLUSION The decrease in absolute values of CD4+ Th cells in severe HA patients may contribute to the establishment of infused FVIII immune tolerance. If the CD4+ Th value remains > 598/µl, clinicians should be vigilant for possible FVIII inhibitor production, especially on days prior to FVIII exposure.
Collapse
Affiliation(s)
- Lu Zhao
- Department of Hematology, Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), Taiyuan 030009, China
| | - Yiqun Zhang
- Department of Hematology, Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), Taiyuan 030009, China
| | - Xinlei Guo
- Department of Hematology, Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), Taiyuan 030009, China
| | - Zhi Li
- Department of Hematology, Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), Taiyuan 030009, China
| | - Wenliang Lu
- Department of Hematology, Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), Taiyuan 030009, China
| | - Zhijuan Pan
- Department of Hematology, Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), Taiyuan 030009, China
| | - Yanru Guo
- Department of Hematology, Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), Taiyuan 030009, China
| | - Jiajia Sun
- Department of Hematology, Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), Taiyuan 030009, China
| | - Ying Zhang
- Department of Hematology, Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), Taiyuan 030009, China
| | - Jinyu Hao
- Department of Hematology, Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), Taiyuan 030009, China
| | - Zhiping Guo
- Department of Hematology, Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), Taiyuan 030009, China
| |
Collapse
|
12
|
Kalia V, Sarkar S. Vitamin D and antiviral immunity. FELDMAN AND PIKE'S VITAMIN D 2024:1011-1034. [DOI: 10.1016/b978-0-323-91338-6.00045-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
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
|
Matveev VA, Mihelic EZ, Benko E, Budylowski P, Grocott S, Lee T, Korosec CS, Colwill K, Stephenson H, Law R, Ward LA, Sheikh-Mohamed S, Mailhot G, Delgado-Brand M, Pasculescu A, Wang JH, Qi F, Tursun T, Kardava L, Chau S, Samaan P, Imran A, Copertino DC, Chao G, Choi Y, Reinhard RJ, Kaul R, Heffernan JM, Jones RB, Chun TW, Moir S, Singer J, Gommerman J, Gingras AC, Kovacs C, Ostrowski M. Immunogenicity of COVID-19 vaccines and their effect on HIV reservoir in older people with HIV. iScience 2023; 26:107915. [PMID: 37790281 PMCID: PMC10542941 DOI: 10.1016/j.isci.2023.107915] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/31/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023] Open
Abstract
Older individuals and people with HIV (PWH) were prioritized for COVID-19 vaccination, yet comprehensive studies of the immunogenicity of these vaccines and their effects on HIV reservoirs are not available. Our study on 68 PWH and 23 HIV-negative participants aged 55 and older post-three vaccine doses showed equally strong anti-spike IgG responses in serum and saliva through week 48 from baseline, while PWH salivary IgA responses were low. PWH had diminished live-virus neutralization responses after two vaccine doses, which were 'rescued' post-booster. Spike-specific T cell immunity was enhanced in PWH with normal CD4+ T cell count, suggesting Th1 imprinting. The frequency of detectable HIV viremia increased post-vaccination, but vaccines did not affect the size of the HIV reservoir in most PWH, except those with low-level viremia. Thus, older PWH require three doses of COVID-19 vaccine for maximum protection, while individuals with unsuppressed viremia should be monitored for adverse reactions from HIV reservoirs.
Collapse
Affiliation(s)
- Vitaliy A. Matveev
- Department of Medicine, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Erik Z. Mihelic
- Department of Medicine, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Erika Benko
- Maple Leaf Medical Clinic, Toronto ON M5G 1K2, Canada
| | - Patrick Budylowski
- Department of Medicine, University of Toronto, Toronto ON M5S 1A8, Canada
- Institute of Medical Science, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Sebastian Grocott
- Department of Medicine, University of Toronto, Toronto ON M5S 1A8, Canada
- Department of Microbiology and Immunology, McGill University, Montreal QC H3A 2B4, Canada
| | - Terry Lee
- CIHR Canadian HIV Trials Network (CTN), Vancouver BC V6Z 1Y6, Canada
- Centre for Health Evaluation and Outcome Sciences (CHÉOS), Vancouver BC V6Z IY6, Canada
| | - Chapin S. Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics Department, York University, Toronto ON M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics Department, York University, Toronto ON M3J 1P3, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto ON M5G 1X5, Canada
| | - Henry Stephenson
- Department of Medicine, University of Toronto, Toronto ON M5S 1A8, Canada
- Department of Bioengineering, McGill University, Montreal QC H3A 0E9, Canada
| | - Ryan Law
- Department of Immunology, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Lesley A. Ward
- Department of Immunology, University of Toronto, Toronto ON M5S 1A8, Canada
| | | | - Geneviève Mailhot
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto ON M5G 1X5, Canada
| | | | - Adrian Pasculescu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto ON M5G 1X5, Canada
| | - Jenny H. Wang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto ON M5G 1X5, Canada
| | - Freda Qi
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto ON M5G 1X5, Canada
| | - Tulunay Tursun
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto ON M5G 1X5, Canada
| | - Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Serena Chau
- Department of Medicine, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Philip Samaan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Annam Imran
- Department of Medicine, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Dennis C. Copertino
- Infectious Diseases, Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Gary Chao
- Department of Immunology, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Yoojin Choi
- Department of Immunology, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Robert J. Reinhard
- Independent Public/Global Health Consultant, San Francisco, CA 94114, USA
| | - Rupert Kaul
- Department of Immunology, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Jane M. Heffernan
- Modelling Infection and Immunity Lab, Mathematics and Statistics Department, York University, Toronto ON M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics Department, York University, Toronto ON M3J 1P3, Canada
| | - R. Brad Jones
- Infectious Diseases, Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joel Singer
- CIHR Canadian HIV Trials Network (CTN), Vancouver BC V6Z 1Y6, Canada
- Centre for Health Evaluation and Outcome Sciences (CHÉOS), Vancouver BC V6Z IY6, Canada
- School of Population and Public Health, University of British Columbia, Vancouver BC V6T 1Z3, Canada
| | - Jennifer Gommerman
- Department of Immunology, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Colin Kovacs
- Maple Leaf Medical Clinic, Toronto ON M5G 1K2, Canada
- Department of Internal Medicine, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Mario Ostrowski
- Department of Medicine, University of Toronto, Toronto ON M5S 1A8, Canada
- Department of Immunology, University of Toronto, Toronto ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health, Toronto ON M5B 1W8, Canada
| |
Collapse
|
15
|
Feng L, Chen X, Huang Y, Zhang X, Zheng S, Xie N. Immunometabolism changes in fibrosis: from mechanisms to therapeutic strategies. Front Pharmacol 2023; 14:1243675. [PMID: 37576819 PMCID: PMC10412938 DOI: 10.3389/fphar.2023.1243675] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Immune cells are essential for initiating and developing the fibrotic process by releasing cytokines and growth factors that activate fibroblasts and promote extracellular matrix deposition. Immunometabolism describes how metabolic alterations affect the function of immune cells and how inflammation and immune responses regulate systemic metabolism. The disturbed immune cell function and their interactions with other cells in the tissue microenvironment lead to the origin and advancement of fibrosis. Understanding the dysregulated metabolic alterations and interactions between fibroblasts and the immune cells is critical for providing new therapeutic targets for fibrosis. This review provides an overview of recent advances in the pathophysiology of fibrosis from the immunometabolism aspect, highlighting the altered metabolic pathways in critical immune cell populations and the impact of inflammation on fibroblast metabolism during the development of fibrosis. We also discuss how this knowledge could be leveraged to develop novel therapeutic strategies for treating fibrotic diseases.
Collapse
Affiliation(s)
- Lixiang Feng
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xingyu Chen
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yujing Huang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xiaodian Zhang
- Hainan Cancer Clinical Medical Center of the First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province and Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Shaojiang Zheng
- Hainan Cancer Clinical Medical Center of the First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province and Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- Department of Pathology, Hainan Women and Children Medical Center, Hainan Medical University, Haikou, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
16
|
Verheul MK, Nijhof KH, de Zeeuw-Brouwer ML, Duijm G, Ten Hulscher H, de Rond L, Beckers L, Eggink D, van Tol S, Reimerink J, Boer M, van Beek J, Rots N, van Binnendijk R, Buisman AM. Booster Immunization Improves Memory B Cell Responses in Older Adults Unresponsive to Primary SARS-CoV-2 Immunization. Vaccines (Basel) 2023; 11:1196. [PMID: 37515012 PMCID: PMC10384172 DOI: 10.3390/vaccines11071196] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
The generation of a specific long-term immune response to SARS-CoV-2 is considered important for protection against COVID-19 infection and disease. Memory B cells, responsible for the generation of antibody-producing plasmablasts upon a new antigen encounter, play an important role in this process. Therefore, the induction of memory B cell responses after primary and booster SARS-CoV-2 immunizations was investigated in the general population with an emphasis on older adults. Participants, 20-99 years of age, due to receive the mRNA-1273 or BNT162b2 SARS-CoV-2 vaccine were included in the current study. Specific memory B cells were determined by ex vivo ELISpot assays. In a subset of participants, antibody levels, avidity, and virus neutralization capacity were compared to memory B cell responses. Memory B cells specific for both Spike S1 and receptor-binding domain (RBD) were detected in the majority of participants following the primary immunization series. However, a proportion of predominantly older adults showed low frequencies of specific memory B cells. Booster vaccination resulted in a large increase in the frequencies of S1- and RBD-specific memory B cells also for those in which low memory B cell frequencies were detected after the primary series. These data show that booster immunization is important for the generation of a memory B cell response, as a subset of older adults shows a suboptimal response to the primary SARS-CoV-2 immunization series. It is anticipated that these memory B cells will play a significant role in the immune response following viral re-exposure.
Collapse
Affiliation(s)
- Marije K Verheul
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Kim H Nijhof
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Mary-Lène de Zeeuw-Brouwer
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Geraly Duijm
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Hinke Ten Hulscher
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Lia de Rond
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Lisa Beckers
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Dirk Eggink
- Centre for Infectious Diseases Research, Diagnostics and Laboratory Surveillance, WHO COVID-19 Reference Laboratory, Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| | - Sophie van Tol
- Centre for Infectious Diseases Research, Diagnostics and Laboratory Surveillance, WHO COVID-19 Reference Laboratory, Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| | - Johan Reimerink
- Centre for Infectious Diseases Research, Diagnostics and Laboratory Surveillance, WHO COVID-19 Reference Laboratory, Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| | - Mardi Boer
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Josine van Beek
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Nynke Rots
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Rob van Binnendijk
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Anne-Marie Buisman
- Centre for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| |
Collapse
|
17
|
Prior JT, Limbert VM, Horowitz RM, D'Souza SJ, Bachnak L, Godwin MS, Bauer DL, Harrell JE, Morici LA, Taylor JJ, McLachlan JB. Establishment of isotype-switched, antigen-specific B cells in multiple mucosal tissues using non-mucosal immunization. NPJ Vaccines 2023; 8:80. [PMID: 37258506 PMCID: PMC10231862 DOI: 10.1038/s41541-023-00677-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
Although most pathogens infect the human body via mucosal surfaces, very few injectable vaccines can specifically target immune cells to these tissues where their effector functions would be most desirable. We have previously shown that certain adjuvants can program vaccine-specific helper T cells to migrate to the gut, even when the vaccine is delivered non-mucosally. It is not known whether this is true for antigen-specific B cell responses. Here we show that a single intradermal vaccination with the adjuvant double mutant heat-labile toxin (dmLT) induces a robust endogenous, vaccine-specific, isotype-switched B cell response. When the vaccine was intradermally boosted, we detected non-circulating vaccine-specific B cell responses in the lamina propria of the large intestines, Peyer's patches, and lungs. When compared to the TLR9 ligand adjuvant CpG, only dmLT was able to drive the establishment of isotype-switched resident B cells in these mucosal tissues, even when the dmLT-adjuvanted vaccine was administered non-mucosally. Further, we found that the transcription factor Batf3 was important for the full germinal center reaction, isotype switching, and Peyer's patch migration of these B cells. Collectively, these data indicate that specific adjuvants can promote mucosal homing and the establishment of activated, antigen-specific B cells in mucosal tissues, even when these adjuvants are delivered by a non-mucosal route. These findings could fundamentally change the way future vaccines are formulated and delivered.
Collapse
Affiliation(s)
- John T Prior
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Vanessa M Limbert
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Rebecca M Horowitz
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Shaina J D'Souza
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Louay Bachnak
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Matthew S Godwin
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - David L Bauer
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jaikin E Harrell
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Lisa A Morici
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Justin J Taylor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - James B McLachlan
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA.
| |
Collapse
|
18
|
Furlong E, Kotecha RS. Lessons learnt from influenza vaccination in immunocompromised children undergoing treatment for cancer. THE LANCET. CHILD & ADOLESCENT HEALTH 2023; 7:199-213. [PMID: 36706776 DOI: 10.1016/s2352-4642(22)00315-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 01/26/2023]
Abstract
Influenza infection contributes substantially to global morbidity and mortality, with children undergoing treatment for cancer among the most vulnerable due to immunosuppression associated with disease and treatment. However, influenza remains one of the most common vaccine-preventable diseases. Despite international guidelines recommending inactivated influenza vaccination on the basis of data supporting efficacy and an excellent safety profile in this population, uptake has often been suboptimal due to persisting hesitancy among both patients and oncologists regarding the ability of the vaccine to mount a sufficient immune response, the optimal vaccine schedule and timing, and the best method to assess response in immunocompromised populations. In this Review, we discuss the evidence regarding influenza vaccination in children with cancer, factors that influence response, and highlight strategies to optimise vaccination. Host immune factors play a substantial role, thus principles learnt from influenza vaccination can be broadly applied for the use of inactivated vaccines in children with cancer.
Collapse
Affiliation(s)
- Eliska Furlong
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, WA, Australia; Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Rishi S Kotecha
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, WA, Australia; Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia; Curtin Medical School, Curtin University, Perth, WA, Australia.
| |
Collapse
|
19
|
Scharf S, Ackermann J, Bender L, Wurzel P, Schäfer H, Hansmann ML, Koch I. Holistic View on the Structure of Immune Response: Petri Net Model. Biomedicines 2023; 11:452. [PMID: 36830988 PMCID: PMC9953182 DOI: 10.3390/biomedicines11020452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/08/2023] Open
Abstract
The simulation of immune response is a challenging task because quantitative data are scarce. Quantitative theoretical models either focus on specific cell-cell interactions or have to make assumptions about parameters. The broad variation of, e.g., the dimensions and abundance between lymph nodes as well as between individual patients hampers conclusive quantitative modeling. No theoretical model has been established representing a consensus on the set of major cellular processes involved in the immune response. In this paper, we apply the Petri net formalism to construct a semi-quantitative mathematical model of the lymph nodes. The model covers the major cellular processes of immune response and fulfills the formal requirements of Petri net models. The intention is to develop a model taking into account the viewpoints of experienced pathologists and computer scientists in the field of systems biology. In order to verify formal requirements, we discuss invariant properties and apply the asynchronous firing rule of a place/transition net. Twenty-five transition invariants cover the model, and each is assigned to a functional mode of the immune response. In simulations, the Petri net model describes the dynamic modes of the immune response, its adaption to antigens, and its loss of memory.
Collapse
Affiliation(s)
- Sonja Scharf
- Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt, Robert-Mayer Str. 11-15, 60325 Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Str. 1, 60438 Frankfurt am Main, Germany
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jörg Ackermann
- Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt, Robert-Mayer Str. 11-15, 60325 Frankfurt am Main, Germany
| | - Leonie Bender
- Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt, Robert-Mayer Str. 11-15, 60325 Frankfurt am Main, Germany
| | - Patrick Wurzel
- Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt, Robert-Mayer Str. 11-15, 60325 Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Str. 1, 60438 Frankfurt am Main, Germany
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Hendrik Schäfer
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Institute of Pathology, Corporate Member of Free University of Berlin, Humboldt-University of Berlin, Charity-University Medicine Berlin, Virchowweg 15, 10117 Berlin, Germany
| | - Martin-Leo Hansmann
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Str. 1, 60438 Frankfurt am Main, Germany
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Ina Koch
- Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt, Robert-Mayer Str. 11-15, 60325 Frankfurt am Main, Germany
| |
Collapse
|
20
|
Guo Z, Wang L, Liu H, Xie Y. Innate Immune Memory in Monocytes and Macrophages: The Potential Therapeutic Strategies for Atherosclerosis. Cells 2022; 11:4072. [PMID: 36552836 PMCID: PMC9776628 DOI: 10.3390/cells11244072] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Atherosclerosis is a complex metabolic disease characterized by the dysfunction of lipid metabolism and chronic inflammation in the intimal space of the vessel. As the most abundant innate immune cells, monocyte-derived macrophages play a pivotal role in the inflammatory response, cholesterol metabolism, and foam cell formation. In recent decades, it has been demonstrated that monocytes and macrophages can establish innate immune memory (also termed trained immunity) via endogenous and exogenous atherogenic stimuli and exhibit a long-lasting proinflammatory phenotype. The important cellular metabolism processes, including glycolysis, oxidative phosphorylation (OXPHOS), the tricarboxylic acid (TCA) cycle, fatty acid synthesis, and cholesterol synthesis, are reprogrammed. Trained monocytes/macrophages with innate immune memory can be persistently hyperactivated and can undergo extensive epigenetic rewiring, which contributes to the pathophysiological development of atherosclerosis via increased proinflammatory cytokine production and lipid accumulation. Here, we provide an overview of the regulation of cellular metabolic processes and epigenetic modifications of innate immune memory in monocytes/macrophages as well as the potential endogenous and exogenous stimulations involved in the progression of atherosclerosis that have been reported recently. These elucidations might be beneficial for further understanding innate immune memory and the development of therapeutic strategies for inflammatory diseases and atherosclerosis.
Collapse
Affiliation(s)
- Zhigang Guo
- Huanghe Science and Technology College, Zhengzhou 450006, China
| | - Lixue Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an 271018, China
| | - Hongjian Liu
- Department of Pharmacy, The Second Affiliated Hospital of Shandong First Medical University, Tai’an 271000, China
| | - Yuhuai Xie
- Huanghe Science and Technology College, Zhengzhou 450006, China
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
21
|
Korosec CS, Farhang-Sardroodi S, Dick DW, Gholami S, Ghaemi MS, Moyles IR, Craig M, Ooi HK, Heffernan JM. Long-term durability of immune responses to the BNT162b2 and mRNA-1273 vaccines based on dosage, age and sex. Sci Rep 2022; 12:21232. [PMID: 36481777 PMCID: PMC9732004 DOI: 10.1038/s41598-022-25134-0] [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: 06/05/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
The lipid nanoparticle (LNP)-formulated mRNA vaccines BNT162b2 and mRNA-1273 are a widely adopted multi vaccination public health strategy to manage the COVID-19 pandemic. Clinical trial data has described the immunogenicity of the vaccine, albeit within a limited study time frame. Here, we use a within-host mathematical model for LNP-formulated mRNA vaccines, informed by available clinical trial data from 2020 to September 2021, to project a longer term understanding of immunity as a function of vaccine type, dosage amount, age, and sex. We estimate that two standard doses of either mRNA-1273 or BNT162b2, with dosage times separated by the company-mandated intervals, results in individuals losing more than 99% humoral immunity relative to peak immunity by 8 months following the second dose. We predict that within an 8 month period following dose two (corresponding to the original CDC time-frame for administration of a third dose), there exists a period of time longer than 1 month where an individual has lost more than 99% humoral immunity relative to peak immunity, regardless of which vaccine was administered. We further find that age has a strong influence in maintaining humoral immunity; by 8 months following dose two we predict that individuals aged 18-55 have a four-fold humoral advantage compared to aged 56-70 and 70+ individuals. We find that sex has little effect on the immune response and long-term IgG counts. Finally, we find that humoral immunity generated from two low doses of mRNA-1273 decays at a substantially slower rate relative to peak immunity gained compared to two standard doses of either mRNA-1273 or BNT162b2. Our predictions highlight the importance of the recommended third booster dose in order to maintain elevated levels of antibodies.
Collapse
Affiliation(s)
- Chapin S Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
| | - Suzan Farhang-Sardroodi
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Department of Mathematics, University of Manitoba, 186 Dysart Road, Winnipeg, MB, R3T 2N2, Canada
| | - David W Dick
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Sameneh Gholami
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Mohammad Sajjad Ghaemi
- Digital Technologies Research Centre, National Research Council Canada, 222 College Street, Toronto, ON, M5T 3J1, Canada
| | - Iain R Moyles
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Morgan Craig
- Department of Mathematics and Statistics, Université de Montréal & Sainte-Justine University Hospital Research Centre, 3175, ch. Côte Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Hsu Kiang Ooi
- Digital Technologies Research Centre, National Research Council Canada, 222 College Street, Toronto, ON, M5T 3J1, Canada
| | - Jane M Heffernan
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
22
|
Dong Y, Pi X, Bartels-Burgahn F, Saltukoglu D, Liang Z, Yang J, Alt FW, Reth M, Wu H. Structural principles of B cell antigen receptor assembly. Nature 2022; 612:156-161. [PMID: 36228656 PMCID: PMC10499536 DOI: 10.1038/s41586-022-05412-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/05/2022] [Indexed: 12/15/2022]
Abstract
The B cell antigen receptor (BCR) is composed of a membrane-bound class M, D, G, E or A immunoglobulin for antigen recognition1-3 and a disulfide-linked Igα (also known as CD79A) and Igβ (also known as CD79B) heterodimer (Igα/β) that functions as the signalling entity through intracellular immunoreceptor tyrosine-based activation motifs (ITAMs)4,5. The organizing principle of the BCR remains unknown. Here we report cryo-electron microscopy structures of mouse full-length IgM BCR and its Fab-deleted form. At the ectodomain (ECD), the Igα/β heterodimer mainly uses Igα to associate with Cµ3 and Cµ4 domains of one heavy chain (µHC) while leaving the other heavy chain (µHC') unbound. The transmembrane domain (TMD) helices of µHC and µHC' interact with those of the Igα/β heterodimer to form a tight four-helix bundle. The asymmetry at the TMD prevents the recruitment of two Igα/β heterodimers. Notably, the connecting peptide between the ECD and TMD of µHC intervenes in between those of Igα and Igβ to guide TMD assembly through charge complementarity. Weaker but distinct density for the Igβ ITAM nestles next to the TMD, suggesting potential autoinhibition of ITAM phosphorylation. Interfacial analyses suggest that all BCR classes utilize a general organizational architecture. Our studies provide a structural platform for understanding B cell signalling and designing rational therapies against BCR-mediated diseases.
Collapse
Affiliation(s)
- Ying Dong
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Xiong Pi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Frauke Bartels-Burgahn
- Signaling Research Centers BIOSS and CIBSS, Freiburg, Germany
- Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Deniz Saltukoglu
- Signaling Research Centers BIOSS and CIBSS, Freiburg, Germany
- Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Zhuoyi Liang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- HHMI, Boston Children's Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Jianying Yang
- Signaling Research Centers BIOSS and CIBSS, Freiburg, Germany
- Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Frederick W Alt
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- HHMI, Boston Children's Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Michael Reth
- Signaling Research Centers BIOSS and CIBSS, Freiburg, Germany.
- Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany.
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
23
|
Pettini E, Medaglini D, Ciabattini A. Profiling the B cell immune response elicited by vaccination against the respiratory virus SARS-CoV-2. Front Immunol 2022; 13:1058748. [PMID: 36505416 PMCID: PMC9729280 DOI: 10.3389/fimmu.2022.1058748] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
B cells play a fundamental role in host defenses against viral infections. Profiling the B cell response elicited by SARS-CoV-2 vaccination, including the generation and persistence of antigen-specific memory B cells, is essential for improving the knowledge of vaccine immune responsiveness, beyond the antibody response. mRNA-based vaccines have shown to induce a robust class-switched memory B cell response that persists overtime and is boosted by further vaccine administration, suggesting that memory B cells are critical in driving a recall response upon re-exposure to SARS-CoV-2 antigens. Here, we focus on the role of the B cell response in the context of SARS-CoV-2 vaccination, offering an overview of the different technologies that can be used to identify spike-specific B cells, characterize their phenotype using machine learning approaches, measure their capacity to reactivate following antigen encounter, and tracking the maturation of the B cell receptor antigenic affinity.
Collapse
Affiliation(s)
| | | | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
24
|
Lai L, Rouphael N, Xu Y, Kabbani S, Beck A, Sherman A, Anderson EJ, Bellamy A, Weiss J, Cross K, Mulligan MJ. An Oil-in-Water adjuvant significantly increased influenza A/H7N9 split virus Vaccine-Induced circulating follicular helper T (cT FH) cells and antibody responses. Vaccine 2022; 40:7065-7072. [PMID: 36273986 DOI: 10.1016/j.vaccine.2022.09.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 04/09/2022] [Accepted: 09/12/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Unadjuvanted A/H7N9 vaccines are poorly immunogenic. The immune response is improved with the addition of MF59, an oil-in-water adjuvant. However, the cellular immunologic responses of MF59-adjuvanted A/H7N9 vaccine are not fully understood. METHODS 37 participants were vaccinated with 2 doses of 2013 influenza A/H7N9 vaccine (at Days 1 and 21) with or without MF59 and enrolled in an immunology substudy. Responses were assessed at multiple timepoints (Days 0, 8, 21, 29, and 42) for hemagglutination inhibition (HAI) and neutralizing antibody (Neut) assays, memory B cell responses by enzyme-linked ImmunoSpot; circulating follicular helper T cells (cTFH) and CD4 + T cells by intracellular cytokine staining. RESULTS MF59-adjuvanted influenza A/H7N9 vaccine induced significantly higher hemagglutination inhibition (HAI) and neutralizing antibody (Neut) responses when compared to unadjuvanted vaccine. The adjuvanted vaccine elicited significantly higher levels of Inducible T-cell Co-Stimulator (ICOS) expression by CXCR3+CXCR5+CD4+ cTFH cells, compared to unadjuvanted vaccine. The magnitude of increase in cTFH cells (from baseline to Day 8) and in IL-21 expressing CD154+CD4+ T cells (from baseline to Days 8 and 21) correlated with HAI (at Day 29) and Neut antibody (at Days 8 and 29) titers. The increase in frequency of IL-21 expressing CD154+CD4+T cells (from baseline to Day 21) correlated with memory B cell frequency (at Day 42). CONCLUSION cTFH activation is associated with HAI and Neut responses in recipients of MF59-adjuvanted influenza A/H7N9 vaccine relative to unadjuvanted vaccine. Future studies should focus on optimizing the cTFH response and use cTFH as an early biomarker of serological response to vaccination. This trial was registered at clinicaltrials.gov, trial number NCT01938742.
Collapse
Affiliation(s)
- Lilin Lai
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030.
| | - Yongxian Xu
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| | - Sarah Kabbani
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| | - Allison Beck
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| | - Amy Sherman
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| | - Evan J Anderson
- Departments of Pediatrics and Medicine, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate
| | - Abbie Bellamy
- EMMES Corporation, 401, North Washington Street, Suite 700, Rockville, MD 20850, USA
| | - Julia Weiss
- EMMES Corporation, 401, North Washington Street, Suite 700, Rockville, MD 20850, USA
| | - Kaitlyn Cross
- EMMES Corporation, 401, North Washington Street, Suite 700, Rockville, MD 20850, USA
| | - Mark J Mulligan
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| |
Collapse
|
25
|
Garcia-Dominguez D, Henry C, Ma L, Jani H, Amato NJ, Manning T, Freyn A, Davis H, Hsiao CJ, Li M, Koch H, Elbashir S, DiPiazza A, Carfi A, Edwards D, Bahl K. Altering the mRNA-1273 dosing interval impacts the kinetics, quality, and magnitude of immune responses in mice. Front Immunol 2022; 13:948335. [PMID: 36426367 PMCID: PMC9679967 DOI: 10.3389/fimmu.2022.948335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/13/2022] [Indexed: 12/17/2024] Open
Abstract
For a vaccine to achieve durable immunity and optimal efficacy, many require a multi-dose primary vaccination schedule that acts to first "prime" naive immune systems and then "boost" initial immune responses by repeated immunizations (ie, prime-boost regimens). In the context of the global coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), 2-dose primary vaccination regimens were often selected with short intervals between doses to provide rapid protection while still inducing robust immunity. However, emerging post-authorization evidence has suggested that longer intervals between doses 1 and 2 for SARS-CoV-2 vaccines may positively impact robustness and durability of immune responses. Here, the dosing interval for mRNA-1273, a messenger RNA based SARS-CoV-2 vaccine administered on a 2-dose primary schedule with 4 weeks between doses, was evaluated in mice by varying the dose interval between 1 and 8 weeks and examining immune responses through 24 weeks after dose 2. A dosing interval of 6 to 8 weeks generated the highest level of antigen-specific serum immunoglobulin G binding antibody titers. Differences in binding antibody titers between mRNA-1273 1 µg and 10 µg decreased over time for dosing intervals of ≥4 weeks, suggesting a potential dose-sparing effect. Longer intervals (≥4 weeks) also increased antibody-dependent cellular cytotoxicity activity and numbers of antibody-secreting cells (including long-lived plasma cells) after the second dose. An interval of 6 to 8 weeks elicited the strongest CD8+ T-cell responses, while an interval of 3 weeks elicited the strongest CD4+ T-cell response. Overall, these results suggest that in a non-pandemic setting, a longer interval (≥6 weeks) between the doses of the primary series for mRNA-1273 may induce more durable immune responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Kapil Bahl
- Moderna, Inc., Cambridge, MA, United States
| |
Collapse
|
26
|
Cheng ZJ, Huang H, Liu Q, Zhong R, Liang Z, Xue M, Liu M, Li S, Wang H, Zheng P, Zheng C, Sun B. Immunoassay and mass cytometry revealed immunological profiles induced by inactivated BBIBP COVID-19 vaccine. J Med Virol 2022; 94:5206-5216. [PMID: 35801663 PMCID: PMC9350407 DOI: 10.1002/jmv.27983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022]
Abstract
With the global prevalence of COVID-19 and the constant emergence of viral variants, boosters for COVID-19 vaccines to enhance antibody titers in human bodies will become an inevitable trend. However, there is a lack of data on antibody levels and the protective effects of booster injections. This study monitored and analyzed the antibody potency and the antibody responses induced by the booster injection in the subjects who received three vaccine doses. The study was conducted in a multicenter collaboration and recruited 360 healthy adults aged 20-74. Participants received the first, second, and booster doses of inactivated Sinopharm/BBIBP COVID-19 vaccine at 0, 1, and 7 months. Vaccine-induced virus-specific antibody levels (SARS-COV-2-IgA/IgM/IgG) were monitored at multiple time points, surrogate virus neutralization test (sVNT), and the spatial distribution and proportion of immune cells and markers were analyzed using the CyTOF method before vaccination and a month after the second dose. The titers of SARS-CoV-2-IgA/IgM/IgG and neutralizing antibodies increased to a high level in the first month after receiving the second dose of vaccine and declined slowly after that. The antibody levels of SARS-CoV-2-IgG and sVNT were significantly increased at 0.5 months after the induction of the booster (p < 0.05). Despite a downward trend, the antibody levels were still high in the following 6 months. The B cell concentration (in humoral sample) a month after the second injection was significantly reduced compared to that before the vaccine injection (p < 0.05). The proportion of the C01 cell cluster was significantly decreased compared with that before vaccine injection (p < 0.05). Individual cell surface markers showed distinctions in spatial distribution but were not significantly different. This study has shown that serum antibody titer levels will decrease with time by monitoring and analyzing the antibody efficacy and the antibody reaction caused by the booster injection of healthy people who received the whole vaccination (completed three injections). Still, the significant peak of the antibody titer levels after booster highlights the recall immune response. It can maintain a high concentration of antibody levels for a long time, which signifies that the protection ability has been enhanced following the injection of booster immunization. Additionally, CyTOF data shows the active production of antibodies and the change in the immunity environment.
Collapse
Affiliation(s)
- Zhangkai J. Cheng
- Department of Allergy and Clinical Immunology, Department of Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina,Medical CollegeInner Mongolia Minzu UniversityTongliaoChina,Guangzhou LaboratoryGuangzhouChina
| | - Huimin Huang
- Department of Allergy and Clinical Immunology, Department of Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Qiwen Liu
- Department of Allergy and Clinical Immunology, Department of Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina,Nanshan SchoolGuangzhou Medical UniversityGuangzhouChina
| | | | - Zhiman Liang
- Department of Allergy and Clinical Immunology, Department of Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Mingshan Xue
- Department of Allergy and Clinical Immunology, Department of Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina,Guangzhou LaboratoryGuangzhouChina
| | - Mingtao Liu
- Department of Allergy and Clinical Immunology, Department of Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Siping Li
- Dongguan Eighth People's HospitalDongguanChina
| | - Hongman Wang
- Fifth Affiliated Hospital of Zunyi Medical UniversityZhuhaiChina
| | - Peiyan Zheng
- Department of Allergy and Clinical Immunology, Department of Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Chunfu Zheng
- Medical CollegeInner Mongolia Minzu UniversityTongliaoChina,Key Laboratory of Zoonose Prevention and ControlUniversities of Inner Mongolia Autonomous RegionTongliaoChina,Department of Microbiology, Immunology and Infectious DiseasesUniversity of CalgaryCalgaryAlbertaCanada
| | - Baoqing Sun
- Department of Allergy and Clinical Immunology, Department of Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina,Guangzhou LaboratoryGuangzhouChina
| |
Collapse
|
27
|
Ma X, Zhu Y, Dong D, Chen Y, Wang S, Yang D, Ma Z, Zhang A, Zhang F, Guo C, Huang Z. Cryo-EM structures of two human B cell receptor isotypes. Science 2022; 377:880-885. [PMID: 35981028 DOI: 10.1126/science.abo3828] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The B cell receptor (BCR) complex plays a critical role in B cell development and immune responses. The assembly mechanisms underlying the BCR complex remain unknown. We determined the cryo-electron microscopy (cryo-EM) structures of human IgG-BCR and IgM-BCR, which consist of membrane-bound immunoglobulin molecules (mIg) and Igα/β subunits at a 1:1 stoichiometry. Assembly of both BCR complexes involves their extracellular domains, membrane-proximal connection peptides, and transmembrane (TM) helices. The TM helices of mIgG and mIgM share a conserved set of hydrophobic and polar interactions with Igα/β TM helices. By contrast, the IgG-Cγ3 and IgM-Cμ4 domains interact with extracellular Ig-like domains of Igα/β through head-to-tail and side-by-side modes, respectively. This work reveals the structural basis for BCR assembly and provides insights into BCR triggering.
Collapse
Affiliation(s)
- Xinyu Ma
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yuwei Zhu
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - De Dong
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yan Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Shubo Wang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Dehui Yang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Zhuo Ma
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Anqi Zhang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Fan Zhang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Changyou Guo
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Zhiwei Huang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| |
Collapse
|
28
|
Fernandez NC, Shinoda K. The Role of B Lymphocyte Subsets in Adipose Tissue Development, Metabolism, and Aging. Compr Physiol 2022; 12:4133-4145. [PMID: 35950657 DOI: 10.1002/cphy.c220006] [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] [Indexed: 11/09/2022]
Abstract
Adipose tissue contains resident B lymphocytes (B cells) with varying immune functions and mechanisms, depending on the adipose depot type and location. The heterogeneity of B cells and their functions affect the immunometabolism of the adipose tissue in aging and age-associated metabolic disorders. B cells exist in categorizations of subsets that have developmental or phenotypic differences with varying functionalities. Subsets can be categorized as either protective or pathogenic depending on their secretion profile or involvement in metabolic maintenance. In this article, we summarized recent finding on the B cell heterogeneity and discuss how we can utilize our current knowledge of adipose resident B lymphocytes for potential treatment for age-associated metabolic disorders. © 2022 American Physiological Society. Compr Physiol 12: 1-13, 2022.
Collapse
Affiliation(s)
- Nicole C Fernandez
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kosaku Shinoda
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Division of Endocrinology & Diabetes, Albert Einstein College of Medicine, Bronx, New York, USA
- Fleischer Institute for Diabetes and Metabolism, Bronx, New York, USA
| |
Collapse
|
29
|
Exploring the mechanism of sweetener neohesperidin dihydrochalcone on oral tolerance via a network pharmacology approach combined with vivo and vitro methods. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
30
|
Schulte D, Peng W, Snijder J. Template-Based Assembly of Proteomic Short Reads For De Novo Antibody Sequencing and Repertoire Profiling. Anal Chem 2022; 94:10391-10399. [PMID: 35834437 PMCID: PMC9330293 DOI: 10.1021/acs.analchem.2c01300] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Antibodies can target a vast molecular diversity of antigens.
This
is achieved by generating a complementary diversity of antibody sequences
through somatic recombination and hypermutation. A full understanding
of the antibody repertoire in health and disease therefore requires
dedicated de novo sequencing methods. Next-generation
cDNA sequencing methods have laid the foundation of our current understanding
of the antibody repertoire, but these methods share one major limitation
in that they target the antibody-producing B-cells, rather than the
functional secreted product in bodily fluids. Mass spectrometry-based
methods offer an opportunity to bridge this gap between antibody repertoire
profiling and bulk serological assays, as they can access antibody
sequence information straight from the secreted polypeptide products.
In a step to meeting the challenge of mass spectrometry (MS)-based
antibody sequencing, we present a fast and simple software tool (Stitch)
to map proteomic short reads to user-defined templates with dedicated
features for both monoclonal antibody sequencing and profiling of
polyclonal antibody repertoires. We demonstrate the use of Stitch
by fully reconstructing two monoclonal antibody sequences with >98%
accuracy (including I/L assignment); sequencing a Fab from patient
serum isolated by reversed-phase liquid chromatography (LC) fractionation
against a high background of homologous antibody sequences; sequencing
antibody light chains from the urine of multiple-myeloma patients;
and profiling the IgG repertoire in sera from patients hospitalized
with COVID-19. We demonstrate that Stitch assembles a comprehensive
overview of the antibody sequences that are represented in the dataset
and provides an important first step toward analyzing polyclonal antibodies
and repertoire profiling.
Collapse
Affiliation(s)
- Douwe Schulte
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Weiwei Peng
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Joost Snijder
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
31
|
Cunha C, Koike T, Seki Y, Yamamoto M, Iwashima M. Schnurri 3 promotes Th2 cytokine production during the late phase of T-cell antigen stimulation. Eur J Immunol 2022; 52:1077-1094. [PMID: 35490426 PMCID: PMC9276650 DOI: 10.1002/eji.202149633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 03/22/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022]
Abstract
Th1 and Th2 polarization is determined by the coordination of numerous factors including the affinity and strength of the antigen-receptor interaction, predominant cytokine environment, and costimulatory molecules present. Here, we show that Schnurri (SHN) proteins have distinct roles in Th1 and Th2 polarization. SHN2 was previously found to block the induction of GATA3 and Th2 differentiation. We found that, in contrast to SHN2, SHN3 is critical for IL-4 production and Th2 polarization. Strength of stimulation controls SHN2 and SHN3 expression patterns, where higher doses of antigen receptor stimulation promoted SHN3 expression and IL-4 production, along with repression of SHN2 expression. SHN3-deficient T cells showed a substantial defect in IL-4 production and expression of AP-1 components, particularly c-Jun and Jun B. This loss of early IL-4 production led to reduced GATA3 expression and impaired Th2 differentiation. Together, these findings uncover SHN3 as a novel, critical regulator of Th2 development.
Collapse
Affiliation(s)
- Christina Cunha
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
| | - Toru Koike
- Department of Biology, Faculty of ScienceShizuoka UniversityShizuokaJapan
| | - Yoichi Seki
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Mutsumi Yamamoto
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Makio Iwashima
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| |
Collapse
|
32
|
Dasari V, Beckett K, Horsefield S, Ambalathingal G, Khanna R. A bivalent CMV vaccine formulated with human compatible TLR9 agonist CpG1018 elicits potent cellular and humoral immunity in HLA expressing mice. PLoS Pathog 2022; 18:e1010403. [PMID: 35737741 PMCID: PMC9223316 DOI: 10.1371/journal.ppat.1010403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/26/2022] [Indexed: 11/30/2022] Open
Abstract
There is now convincing evidence that the successful development of an effective CMV vaccine will require improved formulation and adjuvant selection that is capable of inducing both humoral and cellular immune responses. Here, we have designed a novel bivalent subunit vaccine formulation based on CMV-encoded oligomeric glycoprotein B (gB) and polyepitope protein in combination with human compatible TLR9 agonist CpG1018. The polyepitope protein includes multiple minimal HLA class I-restricted CD8+ T cell epitopes from different antigens of CMV. This subunit vaccine generated durable anti-viral antibodies, CMV-specific CD4+ and CD8+ T cell responses in multiple HLA expressing mice. Antibody responses included broad TH1 isotypes (IgG2a, IgG2b and IgG3) and potently neutralized CMV infection in fibroblasts and epithelial cells. Furthermore, polyfunctional antigen-specific T cell immunity and antiviral antibody responses showed long-term memory maintenance. These observations argue that this novel vaccine strategy, if applied to humans, could facilitate the generation of robust humoral and cellular immune responses which may be more effective in preventing CMV-associated complications in various clinical settings. Human Cytomegalovirus (CMV) is a significant human pathogen. Generally, in healthy people CMV causes mild symptomatic disease, but during pregnancy CMV can transmit from mother to foetus (1 out of every 200 live births worldwide) and lead to sensorineural hearing loss, vision impairment and central nervous system damage. In transplant patients, CMV can cause serious complications leading to organ rejection and even death. Currently, there is no licensed vaccine available to prevent CMV-associated complications in pregnant women and transplant patients. Here, we have developed a novel bivalent CMV vaccine formulation consisting of recombinant CMVpoly and gB proteins in combination with human compatible adjuvant CpG1018. Preclinical immunogenicity evaluation in multiple HLA expressing mice demonstrated that bivalent CMV vaccine formulation consistently generated robust CMV-specific neutralising antibodies, CD4+ and CD8+ T cell responses. More importantly, long-term follow-up analysis showed that the CMV vaccine can induce durable CMV-specific humoral and cellular immune responses. Our results support further development of this bivalent subunit CMV vaccine to test safety, immunogenicity and efficacy in humans.
Collapse
Affiliation(s)
- Vijayendra Dasari
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- * E-mail: (VD); (RK)
| | - Kirrilee Beckett
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Shane Horsefield
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - George Ambalathingal
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Rajiv Khanna
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- * E-mail: (VD); (RK)
| |
Collapse
|
33
|
Pape KA, Dileepan T, Matchett WE, Ellwood C, Stresemann S, Kabage AJ, Kozysa D, Evert C, Matson M, Lopez S, Krueger PD, Graiziger CT, Vaughn BP, Shmidt E, Rhein J, Schacker TW, Bold TD, Langlois RA, Khoruts A, Jenkins MK. Boosting corrects a memory B cell defect in SARS-CoV-2 mRNA-vaccinated patients with inflammatory bowel disease. JCI Insight 2022; 7:159618. [PMID: 35730567 PMCID: PMC9309052 DOI: 10.1172/jci.insight.159618] [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: 02/23/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022] Open
Abstract
Immunosuppressed patients with inflammatory bowel disease (IBD) generate lower amounts of SARS-CoV-2 spike antibodies after mRNA vaccination than healthy controls. We assessed SARS-CoV-2 spike S1 receptor binding domain–specific (S1-RBD–specific) B lymphocytes to identify the underlying cellular defects. Patients with IBD produced fewer anti–S1-RBD antibody–secreting B cells than controls after the first mRNA vaccination and lower amounts of total and neutralizing antibodies after the second. S1-RBD–specific memory B cells were generated to the same degree in IBD and control groups and were numerically stable for 5 months. However, the memory B cells in patients with IBD had a lower S1-RBD–binding capacity than those in controls, which is indicative of a defect in antibody affinity maturation. Administration of a third shot to patients with IBD elevated serum antibodies and generated memory B cells with a normal antigen-binding capacity. These results show that patients with IBD have defects in the formation of antibody-secreting B cells and affinity-matured memory B cells that are corrected by a third vaccination.
Collapse
Affiliation(s)
- Kathryn A Pape
- Department of Microbiology and Immunology.,Center for Immunology
| | | | | | - Charles Ellwood
- Department of Microbiology and Immunology.,Center for Immunology
| | | | | | - Daria Kozysa
- Department of Medicine, Division of Gastroenterology, and
| | - Clayton Evert
- Department of Medicine, Division of Gastroenterology, and
| | - Michael Matson
- Department of Medicine, Division of Gastroenterology, and
| | - Sharon Lopez
- Department of Medicine, Division of Gastroenterology, and
| | - Peter D Krueger
- Department of Microbiology and Immunology.,Center for Immunology
| | | | - Byron P Vaughn
- Department of Medicine, Division of Gastroenterology, and
| | - Eugenia Shmidt
- Department of Medicine, Division of Gastroenterology, and
| | - Joshua Rhein
- Department of Medicine, Division of Infectious Disease, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Timothy W Schacker
- Department of Medicine, Division of Infectious Disease, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Tyler D Bold
- Center for Immunology.,Department of Medicine, Division of Infectious Disease, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Ryan A Langlois
- Department of Microbiology and Immunology.,Center for Immunology
| | - Alexander Khoruts
- Center for Immunology.,Department of Medicine, Division of Gastroenterology, and
| | - Marc K Jenkins
- Department of Microbiology and Immunology.,Center for Immunology
| |
Collapse
|
34
|
Yamasaki K, Kaneko E, Mori K. Establishment of Membrane-Bound IgA1-Specific Antibody Possessing Antibody-Dependent Cellular Cytotoxicity Activity. Monoclon Antib Immunodiagn Immunother 2022; 41:125-132. [PMID: 35736627 DOI: 10.1089/mab.2021.0032] [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: 11/13/2022] Open
Abstract
Therapeutic agents targeting B cells, such as rituximab, have been proven to be effective in several diseases. However, since this therapy targets the whole B cell population, there are still concerns about critical adverse events. Therefore, a new type of antibody that can specifically deplete a particular type of B cell may have the potential to become an efficient and safer therapy. Membrane-bound IgA1 (mIgA1) is expressed on soluble IgA1 (sIgA1) producing B cells and/or their precursor, B cells. Although most of the amino acid sequence in the N-terminal regions of the extracellular domains of mIgA1 and sIgA1 is shared, there is a membrane type-specific region, named the membrane-bound immunoglobulin isotype-specific (migis-α) region, at the membrane-proximal region of mIgA1. We hypothesized that the migis-α region would be a suitable antigen for therapeutic antibodies to target mIgA1-expressing B cells without binding to sIgA1, which may cause undesired adverse effects and poor pharmacokinetics (PK). We established two anti-migis-α monoclonal antibodies (mAbs), KM4641 and KM4644, by immunization of the synthetic peptide corresponding to an migis-α region. These mAbs both showed robust binding to mIgA1-expressing transfectant cells. As we expected, neither mAbs bound to sIgA1 and we found that the mAbs recognized different seven to eight amino acid sequences within the migis-α region. Furthermore, the rat-human chimeric type of both mAbs showed antibody-dependent cellular cytotoxicity against mIgA1-expressing transfectant cells. Taken together, this study showed that established mAbs have therapeutic potential in IgA-related diseases, such as IgA nephropathy.
Collapse
|
35
|
Bai Z, Zhou T, Yu Z, Chen Y, Dong L. Clinical value of 18F-FDG PET/CT in IgG4-related disease. Ann Nucl Med 2022; 36:651-660. [PMID: 35604531 DOI: 10.1007/s12149-022-01749-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/27/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the clinical value of 18F-fluoro-2-deoxy-D-glucose positron emission tomography/computed tomography (18F-FDG PET/CT) in IgG4-related disease (IgG4-RD). METHODS Seventy two patients diagnosed with IgG4-RD who underwent PET/CT were included. Correlations between clinical variables and PET/CT findings were analyzed by Spearman's correlation test. Conventional radiology was compared to PET/CT to evaluate detection discrepancies. The detection ability of insidious organ involvement by PET/CT at disease onset was investigated. The utility value of PET/CT for the 2019 ACR/EULAR classification criteria was analyzed with the multivariate logistic analysis and ROC curve. RESULTS SUVmax of main involved organ was positively correlated with IgG4-RD Responder Index (IgG4-RD RI), serum and tissue IgG4 levels and IgG4/IgG ratio, serum eosinophils counts and number of involved organs, while negatively correlated with serum IgM levels. PET/CT was superior in detecting organ/tissue involvements including prostate, gastrointestinal tract and lung compared with conventional imaging. For patients with pancreato-hepato-biliary or head-neck involvements at onset, PET/CT showed superiority in detecting insidious lesions. Multivariate analysis showed that disease duration, multiple-organ involvement, SUVmax of main involved organ and mean SUVmax of all involved organs were significantly associated with the fulfillment of the 2019 ACR/EULAR classification criteria. ROC curves indicated that the cut-off value for SUVmax of main involved organ and mean SUVmax of all involved organs for fulfillment of the 2019 ACR/EULAR classification criteria for IgG4-RD were 4.1 and 3.5, respectively. CONCLUSION 18F-FDG PET/CT has potential capacity to monitor disease activity, evaluate organ involvements and assist in the classification criteria in IgG4-RD.
Collapse
Affiliation(s)
- Zhiqian Bai
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095th Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Tianshu Zhou
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095th Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Zhihua Yu
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu Chen
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095th Jiefang Avenue, Wuhan, 430022, Hubei, China.
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095th Jiefang Avenue, Wuhan, 430022, Hubei, China.
| |
Collapse
|
36
|
D’Avola A, Legrave N, Tajan M, Chakravarty P, Shearer RL, King HW, Kluckova K, Cheung EC, Clear AJ, Gunawan AS, Zhang L, James LK, MacRae JI, Gribben JG, Calado DP, Vousden KH, Riches JC. PHGDH is required for germinal center formation and is a therapeutic target in MYC-driven lymphoma. J Clin Invest 2022; 132:e153436. [PMID: 35316216 PMCID: PMC9057607 DOI: 10.1172/jci153436] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
The synthesis of serine from glucose is a key metabolic pathway supporting cellular proliferation in healthy and malignant cells. Despite this, the role that this aspect of metabolism plays in germinal center biology and pathology is not known. Here, we performed a comprehensive characterization of the role of the serine synthesis pathway in germinal center B cells and lymphomas derived from these cells. We demonstrate that upregulation of a functional serine synthesis pathway is a metabolic hallmark of B cell activation and the germinal center reaction. Inhibition of phosphoglycerate dehydrogenase (PHGDH), the first and rate-limiting enzyme in this pathway, led to defective germinal formation and impaired high-affinity antibody production. In addition, overexpression of enzymes involved in serine synthesis was a characteristic of germinal center B cell-derived lymphomas, with high levels of expression being predictive of reduced overall survival in diffuse large B cell lymphoma. Inhibition of PHGDH induced apoptosis in lymphoma cells, reducing disease progression. These findings establish PHGDH as a critical player in humoral immunity and a clinically relevant target in lymphoma.
Collapse
Affiliation(s)
| | | | - Mylène Tajan
- The Francis Crick Institute, London, United Kingdom
| | | | | | - Hamish W. King
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | | | | | | | | | | | - Louisa K. James
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - James I. MacRae
- Metabolomics Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | | | | | | | - John C. Riches
- The Francis Crick Institute, London, United Kingdom
- Centre for Haemato-Oncology, Barts Cancer Institute, and
| |
Collapse
|
37
|
van den Hoogen LL, Verheul MK, Vos ERA, van Hagen CCE, van Boven M, Wong D, Wijmenga-Monsuur AJ, Smits G, Kuijer M, van Rooijen D, Bogaard-van Maurik M, Zutt I, van Vliet J, Wolf J, van der Klis FRM, de Melker HE, van Binnendijk RS, den Hartog G. SARS-CoV-2 Spike S1-specific IgG kinetic profiles following mRNA or vector-based vaccination in the general Dutch population show distinct kinetics. Sci Rep 2022; 12:5935. [PMID: 35396570 PMCID: PMC8990276 DOI: 10.1038/s41598-022-10020-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/30/2022] [Indexed: 12/21/2022] Open
Abstract
mRNA- and vector-based vaccines are used at a large scale to prevent COVID-19. We compared Spike S1-specific (S1) IgG antibodies after vaccination with mRNA-based (Comirnaty, Spikevax) or vector-based (Janssen, Vaxzevria) vaccines, using samples from a Dutch nationwide cohort. In adults 18-64 years old (n = 2412), the median vaccination interval between the two doses was 77 days for Vaxzevria (interquartile range, IQR: 69-77), 35 days (28-35) for Comirnaty and 33 days (28-35) for Spikevax. mRNA vaccines induced faster inclines and higher S1 antibodies compared to vector-based vaccines. For all vaccines, one dose resulted in boosting of S1 antibodies in adults with a history of SARS-CoV-2 infection. For Comirnaty, two to four months following the second dose (n = 196), S1 antibodies in adults aged 18-64 years old (436 BAU/mL, IQR: 328-891) were less variable and median concentrations higher compared to those in persons ≥ 80 years old (366, 177-743), but differences were not statistically significant (p > 0.100). Nearly all participants seroconverted following COVID-19 vaccination, including the aging population. These data confirm results from controlled vaccine trials in a general population, including vulnerable groups.
Collapse
Affiliation(s)
- Lotus L van den Hoogen
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Marije K Verheul
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Eric R A Vos
- Centre for Infectious Diseases, Epidemiology and Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Cheyenne C E van Hagen
- Centre for Infectious Diseases, Epidemiology and Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Michiel van Boven
- Centre for Infectious Diseases, Epidemiology and Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Denise Wong
- Centre for Infectious Diseases, Epidemiology and Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Alienke J Wijmenga-Monsuur
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Gaby Smits
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Marjan Kuijer
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Debbie van Rooijen
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Marjan Bogaard-van Maurik
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Ilse Zutt
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Jeffrey van Vliet
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Janine Wolf
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Fiona R M van der Klis
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Hester E de Melker
- Centre for Infectious Diseases, Epidemiology and Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Robert S van Binnendijk
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Gerco den Hartog
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands.
| |
Collapse
|
38
|
Dharmaratnam A, Sudhagar A, Das S, Nair RR, Nithianantham SR, Preena PG, Lekshmi N, Swaminathan TR. Immune gene expression and protective effects in goldfish (Carassius auratus L.) immunized with formalin-inactivated cyprinid herpesvirus-2 (CyHV-2) vaccine. Microb Pathog 2022; 164:105452. [PMID: 35217179 DOI: 10.1016/j.micpath.2022.105452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 10/19/2022]
Abstract
The goldfish hematopoietic necrosis viral disease (GHNVD) has led to worldwide economic losses in goldfish aquaculture. The present study has focused on the development of an inactivated vaccine for the cyprinid herpesvirus (CyHV-2) and to check the immunogenicity of the vaccine in the host. The fantail goldfish fin (FtGF) cell line was used in the propagation of the CyHV-2 and the viral titer obtained were of 107.8 TCID50/ml. Followed by the virus was inactivated using 0.1% formalin for 2 days. Various concentrations of formalin-inactivated CyHV-2 (1%, 0.7%, 0.5%, 0.3% and 0.1%) were studied in the FtGF cell line. Morphological changes were observed in the FtGF cell line in all other concentrations of formalin except 0.1% formalin-inactivated CyHV-2 vaccine. The goldfishes were intraperitoneally injected with 300 μl of vaccine and various immune gene responses were studied for a period of 30 days. The gene expression of the adaptive markers CD8, CD4, IFN-ϒ, the cytokines (IL-10, IL-12) was studied in kidney and spleen tissues. Formalin-inactivated CyHV-2 vaccine showed a significant up-regulation of the genes CD8 and IFN-ϒ by the 6th hr post-vaccination onwards. The experimental fish were challenged intraperitoneally with CyHV-2 virus of concentration 107.8 TCID50/ml after 30 days of post-vaccination. A significant difference in cumulative mortality rate was observed for the vaccinated fishes from the unvaccinated fishes. The relative percent survival for formalin immunized fish was 74.03%. Our results have proven that the formalin-inactivated vaccines were efficient and it resulted in triggering the immune gene expression in goldfish. The development and further enhanced studies for this vaccine will lead to a promising low-cost commercial vaccine for CyHV-2 viral infection.
Collapse
Affiliation(s)
- Arathi Dharmaratnam
- PMFGR Centre National Bureau of Fish Genetic Resources, Kochi, Kerala, 682018, India
| | - Arun Sudhagar
- PMFGR Centre National Bureau of Fish Genetic Resources, Kochi, Kerala, 682018, India
| | - Sweta Das
- PMFGR Centre National Bureau of Fish Genetic Resources, Kochi, Kerala, 682018, India
| | - Reshma Rajeev Nair
- PMFGR Centre National Bureau of Fish Genetic Resources, Kochi, Kerala, 682018, India
| | | | | | - Narendrakumar Lekshmi
- PMFGR Centre National Bureau of Fish Genetic Resources, Kochi, Kerala, 682018, India
| | | |
Collapse
|
39
|
Khan RA, Ahmed F, Afroz S, Khan N. Tetravalent formulation of Polymeric nanoparticle-based vaccine induces a potent immune response against Dengue virus. Biomater Sci 2022; 10:2917-2928. [DOI: 10.1039/d2bm00167e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dengue is a mosquito-borne disease caused by the four serotypes of dengue virus (DENV 1-4). It is growing at an alarming rate globally, which could be partly attributed to the...
Collapse
|
40
|
Abstract
The SARS-CoV-2 infection spread rapidly throughout the world and appears to involve in both humoral and cell-mediated immunity. SARS-CoV-2 is attached to host cells via binding to the viral spike (S) proteins and its cellular receptors angiotensin-converting enzyme 2 (ACE2). Consequently, the S protein is primed with serine proteases TMPRSS2 and TMPRSS4, which facilitate the fusion of viral and cellular membranes result in the entry of viral RNA into the host cell. Vaccines are urgently required to combat the coronavirus disease 2019 (COVID-19) outbreak and aid in the recovery to pre-pandemic levels of normality. The long-term protective immunity is provided by the vaccine antigen (or pathogen)-specific immune effectors and the activation of immune memory cells that can be efficiently and rapidly reactivated upon pathogen exposure. Research efforts aimed towards the design and development of vaccines for SARS-CoV-2 are increasing. Numerous coronavirus disease 2019 (COVID-19) vaccines have passed late-stage clinical investigations with promising outcomes. This review focuses on the present state and future prospects of COVID-19 vaccines research and development, with a particular emphasis on immunological mechanisms of various COVID-19vaccines such as adenoviral vector-based vaccines, mRNA vaccines, and DNA vaccines that elicits immunological responses against SARS-CoV-2 infections in humans.
Collapse
|
41
|
Bahroudi M, Bakhshi B, Soudi S, Najar-Peerayeh S. Immunomodulatory effects of mesenchymal stem cell-conditioned media on lipopolysaccharide of Vibrio cholerae as a vaccine candidate. Stem Cell Res Ther 2021; 12:564. [PMID: 34732259 PMCID: PMC8567566 DOI: 10.1186/s13287-021-02622-0] [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: 05/08/2021] [Accepted: 10/11/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Vibrio cholerae is the causative agent of cholera, which is commonly associated with high morbidity and mortality, and presents a major challenge to healthcare systems throughout the world. Lipopolysaccharide (LPS) is required for full protection against V. cholerae but can induce inflammation and septic shock. Mesenchymal stem cells (MSCs) are currently used to treat infectious and inflammatory diseases. Therefore, this study aimed to evaluate the immune-modulating effects of the LPS-MSC-conditioned medium (CM) on V. cholerae LPS immunization in a murine model. METHODS After preconditioning MSCs with LPS, mice were immunized intraperitoneally on days 0 and 14 with the following combinations: LPS + LPS-MSC-CM; detoxified LPS (DLPS) + MSC-CM; LPS + MSC sup; LPS; LPS-MSC-CM; MSC supernatant (MSC sup); and PBS. The mouse serum and saliva samples were collected to evaluate antibody (serum IgG and saliva IgA) and cytokine responses (TNF-α, IL-10, IL-6, TGF-β, IL-4, IL-5, and B-cell activating factor (BAFF)). RESULTS The LPS + LPS-MSC-CM significantly increased total IgG and IgA compared to other combinations (P < 0.001). TNF-α levels, in contrast to IL-10 and TGF-β, were reduced significantly in mice receiving the LPS + LPS-MSC-CM compared to mice receiving only LPS. IL-4, IL-5, and BAFF levels significantly increased in mice receiving increased doses of LPS + LPS-MSC-CM compared to those who received only LPS. The highest vibriocidal antibody titer (1:64) was observed in LPS + LPS-MSC-CM-immunized mice and resulted in a significant improvement in survival in infant mice infected by V. cholerae O1. CONCLUSIONS The LPS-MSC-CM modulates the immune response to V. cholerae LPS by regulating inflammatory and anti-inflammatory responses and inducing vibriocidal antibodies, which protect neonate mice against V. cholerae infection.
Collapse
Affiliation(s)
- Mahboube Bahroudi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Jalal-Ale-Ahmad Ave., 14117-13116, Tehran, Iran
| | - Bita Bakhshi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Jalal-Ale-Ahmad Ave., 14117-13116, Tehran, Iran.
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Jalal-Ale-Ahmad Ave., 14117-13116, Tehran, Iran
| | - Shahin Najar-Peerayeh
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Jalal-Ale-Ahmad Ave., 14117-13116, Tehran, Iran
| |
Collapse
|
42
|
Pape KA, Dileepan T, Kabage AJ, Kozysa D, Batres R, Evert C, Matson M, Lopez S, Krueger PD, Graiziger C, Vaughn BP, Shmidt E, Rhein J, Schacker TW, Khoruts A, Jenkins MK. High-affinity memory B cells induced by SARS-CoV-2 infection produce more plasmablasts and atypical memory B cells than those primed by mRNA vaccines. Cell Rep 2021; 37:109823. [PMID: 34610291 PMCID: PMC8463313 DOI: 10.1016/j.celrep.2021.109823] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/16/2021] [Accepted: 09/20/2021] [Indexed: 02/03/2023] Open
Abstract
Although both infections and vaccines induce memory B cell (MBC) populations that participate in secondary immune responses, the MBCs generated in each case can differ. Here, we compare SARS-CoV-2 spike receptor binding domain (S1-RBD)-specific primary MBCs that form in response to infection or a single mRNA vaccination. Both primary MBC populations have similar frequencies in the blood and respond to a second S1-RBD exposure by rapidly producing plasmablasts with an abundant immunoglobulin (Ig)A+ subset and secondary MBCs that are mostly IgG+ and cross-react with the B.1.351 variant. However, infection-induced primary MBCs have better antigen-binding capacity and generate more plasmablasts and secondary MBCs of the classical and atypical subsets than do vaccine-induced primary MBCs. Our results suggest that infection-induced primary MBCs have undergone more affinity maturation than vaccine-induced primary MBCs and produce more robust secondary responses.
Collapse
Affiliation(s)
- Kathryn A Pape
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Thamotharampillai Dileepan
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Amanda J Kabage
- Department of Medicine, Division of Gastroenterology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Daria Kozysa
- Department of Medicine, Division of Gastroenterology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Rodolfo Batres
- Department of Medicine, Division of Infectious Disease, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Clayton Evert
- Department of Medicine, Division of Gastroenterology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Michael Matson
- Department of Medicine, Division of Gastroenterology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Sharon Lopez
- Department of Medicine, Division of Gastroenterology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Peter D Krueger
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Carolyn Graiziger
- Department of Medicine, Division of Gastroenterology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Byron P Vaughn
- Department of Medicine, Division of Gastroenterology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Eugenia Shmidt
- Department of Medicine, Division of Gastroenterology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Joshua Rhein
- Department of Medicine, Division of Infectious Disease, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Timothy W Schacker
- Department of Medicine, Division of Infectious Disease, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Alexander Khoruts
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Medicine, Division of Gastroenterology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Marc K Jenkins
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
43
|
Czaja AJ. Review article: targeting the B cell activation system in autoimmune hepatitis. Aliment Pharmacol Ther 2021; 54:902-922. [PMID: 34506662 DOI: 10.1111/apt.16574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/30/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The B cell activation system, consisting of B cell activating factor and a proliferation-inducing ligand, may have pathogenic effects in autoimmune hepatitis. AIMS To describe the biological actions of the B cell activation system, indicate its possible role in autoimmune diseases, and evaluate its prospects as a therapeutic target in autoimmune hepatitis METHODS: English abstracts were identified in PubMed by multiple search terms. Full length articles were selected for review, and secondary and tertiary bibliographies were developed. RESULTS The B cell activating factor is crucial for the maturation and survival of B cells, and it can co-stimulate T cell activation, proliferation, and survival. It can also modulate the immune response by inducing interleukin 10 production by regulatory B cells. A proliferation-inducing ligand modulates and diversifies the antibody response by inducing class-switch recombination in B cells. It can also increase the proliferation, survival, and antigen activation of T cells. These immune stimulatory actions can be modulated by inducing proliferation of regulatory T cells. The B cell activation system has been implicated in diverse autoimmune diseases, and therapeutic blockade is a management strategy now being evaluated in autoimmune hepatitis. CONCLUSIONS The B cell activation system has profound effects on B and T cell function in autoimmune diseases. Blockade therapy is being actively evaluated in autoimmune hepatitis. Clarification of the critical pathogenic components of the B cell activation system will improve the targeting, efficacy, and safety of blockade therapy in this disease.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
44
|
Patterson DG, Kania AK, Zuo Z, Scharer CD, Boss JM. Epigenetic gene regulation in plasma cells. Immunol Rev 2021; 303:8-22. [PMID: 34010461 PMCID: PMC8387415 DOI: 10.1111/imr.12975] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022]
Abstract
Humoral immunity provides protection from pathogenic infection and is mediated by antibodies following the differentiation of naive B cells (nBs) to antibody-secreting cells (ASCs). This process requires substantial epigenetic and transcriptional rewiring to ultimately repress the nB program and replace it with one conducive to ASC physiology and function. Notably, these reprogramming events occur within the framework of cell division. Efforts to understand the relationship of cell division with reprogramming and ASC differentiation in vivo have uncovered the timing and scope of reprogramming, as well as key factors that influence these events. Herein, we discuss the unique physiology of ASC and how nBs undergo epigenetic and genome architectural reorganization to acquire the necessary functions to support antibody production. We also discuss the stage-wise manner in which reprogramming occurs across cell divisions and how key molecular determinants can influence B cell fate outcomes.
Collapse
Affiliation(s)
- Dillon G. Patterson
- Department of Microbiology and Immunology, Emory University, Atlanta GA 30322
| | - Anna K. Kania
- Department of Microbiology and Immunology, Emory University, Atlanta GA 30322
| | - Zhihong Zuo
- Department of Microbiology and Immunology, Emory University, Atlanta GA 30322
- Xiangya School of Medicine, Central South University, Changsha, 410008, China
| | | | - Jeremy M. Boss
- Department of Microbiology and Immunology, Emory University, Atlanta GA 30322
| |
Collapse
|
45
|
Blanchard-Rohner G. Vaccination in Children With Autoimmune Disorders and Treated With Various Immunosuppressive Regimens: A Comprehensive Review and Practical Guide. Front Immunol 2021; 12:711637. [PMID: 34408752 PMCID: PMC8365419 DOI: 10.3389/fimmu.2021.711637] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/12/2021] [Indexed: 01/04/2023] Open
Abstract
Children with autoimmune disorders are especially at risk of vaccine-preventable diseases due to their underlying disease and the immunosuppressive treatment often required for a long period. In addition, vaccine coverage remains too low in this vulnerable population. This can be explained by a fear of possible adverse effects of vaccines under immunosuppression, but also a lack of data and clear recommendations, particularly with regard to vaccination with live vaccines. In this review, the latest literature and recommendations on vaccination in immunosuppressed children are discussed in detail, with the aim to provide a set of practical guidelines on vaccination for specialists caring for children suffering from different autoimmune disorders and treated with various immunosuppressive regimens.
Collapse
Affiliation(s)
- Geraldine Blanchard-Rohner
- Paediatric Immunology and Vaccinology Unit, Division of General Paediatrics, Department of Paediatrics, Gynaecology and Obstetrics, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Centre for Vaccinology and Neonatal Immunology, Department of Paediatrics and Pathology-Immunology, Medical Faculty and University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
46
|
Jang A, Sharp R, Wang JM, Feng Y, Wang J, Chen M. Dependence on Autophagy for Autoreactive Memory B Cells in the Development of Pristane-Induced Lupus. Front Immunol 2021; 12:701066. [PMID: 34335611 PMCID: PMC8322733 DOI: 10.3389/fimmu.2021.701066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/30/2021] [Indexed: 02/02/2023] Open
Abstract
The production of autoantibodies by autoreactive B cells plays a major role in the pathogenesis of lupus. Increases in memory B cells have been observed in human lupus patients and autoimmune lpr mice. Autophagy is required for the maintenance of memory B cells against viral infections; however, whether autophagy regulates the persistence of autoantigen-specific memory B cells and the development of lupus remains to be determined. Here we show that memory B cells specific for autoantigens can be detected in autoimmune lpr mice and a pristane-induced lupus mouse model. Interestingly, B cell-specific deletion of Atg7 led to significant loss of autoreactive memory B cells and reduced autoantibody production in pristane-treated mice. Autophagy deficiency also attenuated the development of autoimmune glomerulonephritis and pulmonary inflammation after pristane treatment. Adoptive transfer of wild type autoreactive memory B cells restored autoantibody production in Atg7-deficient recipients. These data suggest that autophagy is important for the persistence of autoreactive memory B cells in mediating autoantibody responses. Our results suggest that autophagy could be targeted to suppress autoreactive memory B cells and ameliorate humoral autoimmunity.
Collapse
Affiliation(s)
- Albert Jang
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Robert Sharp
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Jeffrey M. Wang
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Yin Feng
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Jin Wang
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX, United States,Department of Surgery, Weill Cornell Medical College, Cornell University, New York, NY, United States,*Correspondence: Jin Wang, ; Min Chen,
| | - Min Chen
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States,*Correspondence: Jin Wang, ; Min Chen,
| |
Collapse
|
47
|
Mincham KT, Young JD, Strickland DH. OMIP 076: High-dimensional immunophenotyping of murine T-cell, B-cell, and antibody secreting cell subsets. Cytometry A 2021; 99:888-892. [PMID: 34159723 PMCID: PMC9546025 DOI: 10.1002/cyto.a.24474] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/14/2021] [Accepted: 05/31/2021] [Indexed: 11/21/2022]
Affiliation(s)
- Kyle T Mincham
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Jacob D Young
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Deborah H Strickland
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
48
|
Rana M, La Bella A, Lederman R, Volpe BT, Sherry B, Diamond B. Follicular dendritic cell dysfunction contributes to impaired antigen-specific humoral responses in sepsis-surviving mice. J Clin Invest 2021; 131:146776. [PMID: 33956665 PMCID: PMC8203464 DOI: 10.1172/jci146776] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/29/2021] [Indexed: 12/29/2022] Open
Abstract
Sepsis survivors exhibit impaired responsiveness to antigen (Ag) challenge associated with increased mortality from infection. The contribution of follicular dendritic cells (FDCs) in the impaired humoral response in sepsis-surviving mice is investigated in this study. We demonstrated that mice subjected to sepsis from cecal ligation and puncture (CLP mice) have reduced NP-specific high-affinity class-switched Ig antibodies (Abs) compared with sham-operated control mice following immunization with the T cell-dependent Ag, NP-CGG. NP-specific germinal center (GC) B cells in CLP mice exhibited reduced TNF-α and AID mRNA expression compared with sham-operated mice. CLP mice showed a reduction in FDC clusters, a reduced binding of immune complexes on FDCs, and reduced mRNA expression of CR2, ICAM-1, VCAM-1, FcγRIIB, TNFR1, IKK2, and LTβR compared with sham-operated mice. Adoptive transfer studies showed that there was no B cell-intrinsic defect. In summary, our data suggest that the reduced Ag-specific Ab response in CLP mice is secondary to a disruption in FDC and GC B cell function.
Collapse
Affiliation(s)
- Minakshi Rana
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases
| | - Andrea La Bella
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases
| | - Rivka Lederman
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases
| | | | - Barbara Sherry
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, New York, New York, USA
| | - Betty Diamond
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases
| |
Collapse
|
49
|
Barron SL, Saez J, Owens RM. In Vitro Models for Studying Respiratory Host-Pathogen Interactions. Adv Biol (Weinh) 2021; 5:e2000624. [PMID: 33943040 PMCID: PMC8212094 DOI: 10.1002/adbi.202000624] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/23/2021] [Indexed: 12/22/2022]
Abstract
Respiratory diseases and lower respiratory tract infections are among the leading cause of death worldwide and, especially given the recent severe acute respiratory syndrome coronavirus-2 pandemic, are of high and prevalent socio-economic importance. In vitro models, which accurately represent the lung microenvironment, are of increasing significance given the ethical concerns around animal work and the lack of translation to human disease, as well as the lengthy time to market and the attrition rates associated with clinical trials. This review gives an overview of the biological and immunological components involved in regulating the respiratory epithelium system in health, disease, and infection. The evolution from 2D to 3D cell biology and to more advanced technological integrated models for studying respiratory host-pathogen interactions are reviewed and provide a reference point for understanding the in vitro modeling requirements. Finally, the current limitations and future perspectives for advancing this field are presented.
Collapse
Affiliation(s)
- Sarah L. Barron
- Bioassay Impurities and QualityBiopharmaceuticals DevelopmentR&DAstraZenecaCambridgeCB21 6GPUK
- Department of Chemical Engineering and BiotechnologyPhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Janire Saez
- Department of Chemical Engineering and BiotechnologyPhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Róisín M. Owens
- Department of Chemical Engineering and BiotechnologyPhilippa Fawcett DriveCambridgeCB3 0ASUK
| |
Collapse
|
50
|
Zhan W, Muhuri M, Tai PWL, Gao G. Vectored Immunotherapeutics for Infectious Diseases: Can rAAVs Be The Game Changers for Fighting Transmissible Pathogens? Front Immunol 2021; 12:673699. [PMID: 34046041 PMCID: PMC8144494 DOI: 10.3389/fimmu.2021.673699] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/23/2021] [Indexed: 01/08/2023] Open
Abstract
Conventional vaccinations and immunotherapies have encountered major roadblocks in preventing infectious diseases like HIV, influenza, and malaria. These challenges are due to the high genomic variation and immunomodulatory mechanisms inherent to these diseases. Passive transfer of broadly neutralizing antibodies may offer partial protection, but these treatments require repeated dosing. Some recombinant viral vectors, such as those based on lentiviruses and adeno-associated viruses (AAVs), can confer long-term transgene expression in the host after a single dose. Particularly, recombinant (r)AAVs have emerged as favorable vectors, given their high in vivo transduction efficiency, proven clinical efficacy, and low immunogenicity profiles. Hence, rAAVs are being explored to deliver recombinant antibodies to confer immunity against infections or to diminish the severity of disease. When used as a vaccination vector for the delivery of antigens, rAAVs enable de novo synthesis of foreign proteins with the conformation and topology that resemble those of natural pathogens. However, technical hurdles like pre-existing immunity to the rAAV capsid and production of anti-drug antibodies can reduce the efficacy of rAAV-vectored immunotherapies. This review summarizes rAAV-based prophylactic and therapeutic strategies developed against infectious diseases that are currently being tested in pre-clinical and clinical studies. Technical challenges and potential solutions will also be discussed.
Collapse
Affiliation(s)
- Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Manish Muhuri
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Phillip W. L. Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|