1
|
Lim CC, Lim TS. Profiling the broad antibody diversity of lymphatic filariasis immune antibody repertoire by deep sequencing. Int J Biol Macromol 2025; 290:140037. [PMID: 39828167 DOI: 10.1016/j.ijbiomac.2025.140037] [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: 06/26/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Lymphatic filariasis is caused by infections of thread-like filarial worms, namely Wuchereria bancrofti, Brugia Malayi and Brugia timori. However, in-depth analysis of the antibody repertoire against Lymphatic filariasis is lacking. Using high-throughput sequencing of antibody repertoires, immunome analysis of IgG (LG) and IgM (LM) repertoires were studied. Despite significant differences between LG and LM in V(D)J gene usage, IGHV4-34, IGHV6-1, IGHD3-10 and IGHJ4 were preferred in both repertoires. The CDR3 in the LG repertoire showed a longer length than LM. Higher SHM level were observed in LG sequences and presence of oligoclonal sequences indicates the extent of clonal expansion. The prevalence of rare clonotypes in LM repertoire depicts the high clonal diversity when compared to LG repertoire. Monoclonal antibodies against closely related parasitic infections were present within the LG repertoire suggesting that immune repertoires may not be as exclusive and biased against the target infection as initially thought. The characterization of the immune repertoire can provide critical insight into the antibody response patterns in disease state, antibody generation process during infections and future antibody designs.
Collapse
Affiliation(s)
- Chia Chiu Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia; Analytical Biochemistry Research Centre, Universiti Sains Malaysia, 11800 Penang, Malaysia.
| |
Collapse
|
2
|
Hao Q, Li J, Yeap LS. Molecular mechanisms of DNA lesion and repair during antibody somatic hypermutation. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2344-2353. [PMID: 39048716 DOI: 10.1007/s11427-024-2615-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/08/2024] [Indexed: 07/27/2024]
Abstract
Antibody diversification is essential for an effective immune response, with somatic hypermutation (SHM) serving as a key molecular process in this adaptation. Activation-induced cytidine deaminase (AID) initiates SHM by inducing DNA lesions, which are ultimately resolved into point mutations, as well as small insertions and deletions (indels). These mutational outcomes contribute to antibody affinity maturation. The mechanisms responsible for generating point mutations and indels involve the base excision repair (BER) and mismatch repair (MMR) pathways, which are well coordinated to maintain genomic integrity while allowing for beneficial mutations to occur. In this regard, translesion synthesis (TLS) polymerases contribute to the diversity of mutational outcomes in antibody genes by enabling the bypass of DNA lesions. This review summarizes our current understanding of the distinct molecular mechanisms that generate point mutations and indels during SHM. Understanding these mechanisms is critical for elucidating the development of broadly neutralizing antibodies (bnAbs) and autoantibodies, and has implications for vaccine design and therapeutics.
Collapse
Affiliation(s)
- Qian Hao
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinfeng Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Leng-Siew Yeap
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
3
|
Tsay GJ, Zouali M. Cellular pathways and molecular events that shape autoantibody production in COVID-19. J Autoimmun 2024; 147:103276. [PMID: 38936147 DOI: 10.1016/j.jaut.2024.103276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/26/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
A hallmark of COVID-19 is the variety of complications that follow SARS-CoV-2 infection in some patients, and that target multiple organs and tissues. Also remarkable are the associations with several auto-inflammatory disorders and the presence of autoantibodies directed to a vast array of antigens. The processes underlying autoantibody production in COVID-19 have not been completed deciphered. Here, we review mechanisms involved in autoantibody production in COVID-19, multisystem inflammatory syndrome in children, and post-acute sequelae of COVID19. We critically discuss how genomic integrity, loss of B cell tolerance to self, superantigen effects of the virus, and extrafollicular B cell activation could underly autoantibody proaction in COVID-19. We also offer models that may account for the pathogenic roles of autoantibodies in the promotion of inflammatory cascades, thromboembolic phenomena, and endothelial and vascular deregulations.
Collapse
Affiliation(s)
- Gregory J Tsay
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan; College of Medicine, China Medical University, Taichung, Taiwan
| | - Moncef Zouali
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
| |
Collapse
|
4
|
Guo J, Chowdhury RR, Mallajosyula V, Xie J, Dubey M, Liu Y, Li J, Wei YL, Palanski BA, Wang C, Qiu L, Ohanyan M, Kask O, Sola E, Kamalyan L, Lewis DB, Scriba TJ, Davis MM, Dodd D, Zeng X, Chien YH. γδ T cell antigen receptor polyspecificity enables T cell responses to a broad range of immune challenges. Proc Natl Acad Sci U S A 2024; 121:e2315592121. [PMID: 38227652 PMCID: PMC10823224 DOI: 10.1073/pnas.2315592121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/22/2023] [Indexed: 01/18/2024] Open
Abstract
γδ T cells are essential for immune defense and modulating physiological processes. While they have the potential to recognize large numbers of antigens through somatic gene rearrangement, the antigens which trigger most γδ T cell response remain unidentified, and the role of antigen recognition in γδ T cell function is contentious. Here, we show that some γδ T cell receptors (TCRs) exhibit polyspecificity, recognizing multiple ligands of diverse molecular nature. These ligands include haptens, metabolites, neurotransmitters, posttranslational modifications, as well as peptides and proteins of microbial and host origin. Polyspecific γδ T cells are enriched among activated cells in naive mice and the responding population in infection. They express diverse TCR sequences, have different functional potentials, and include the innate-like γδ T cells, such as the major IL-17 responders in various pathological/physiological conditions. We demonstrate that encountering their antigenic microbiome metabolite maintains their homeostasis and functional response, indicating that their ability to recognize multiple ligands is essential for their function. Human γδ T cells with similar polyspecificity also respond to various immune challenges. This study demonstrates that polyspecificity is a prevalent feature of γδ T cell antigen recognition, which enables rapid and robust T cell responses to a wide range of challenges, highlighting a unique function of γδ T cells.
Collapse
Affiliation(s)
- Jing Guo
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Roshni Roy Chowdhury
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Vamsee Mallajosyula
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA94305
| | - Jianming Xie
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Megha Dubey
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Yuanyuan Liu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Jing Li
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA94305
| | - Yu-ling Wei
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | | | - Conghua Wang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Lingfeng Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, China
- National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, China
| | - Mané Ohanyan
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Oliver Kask
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Elsa Sola
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA94305
| | - Lilit Kamalyan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA94305
| | - David B. Lewis
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA94305
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town7700, South Africa
| | - Mark M. Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA94305
- HHMI, Stanford University School of Medicine, Stanford, CA94305
| | - Dylan Dodd
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Xun Zeng
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, China
- National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, China
- Research Units of Infectious disease and Microecology, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Yueh-hsiu Chien
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
5
|
Leontieva G, Gupalova T, Desheva Y, Kramskaya T, Bormotova E, Koroleva I, Kopteva O, Suvorov A. Evaluation of Immune Response to Mucosal Immunization with an Oral Probiotic-Based Vaccine in Mice: Potential for Prime-Boost Immunization against SARS-CoV-2. Int J Mol Sci 2023; 25:215. [PMID: 38203387 PMCID: PMC10779021 DOI: 10.3390/ijms25010215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/06/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Following the conclusion of the COVID-19 pandemic, the persistent genetic variability in the virus and its ongoing circulation within the global population necessitate the enhancement of existing preventive vaccines and the development of novel ones. A while back, we engineered an orally administered probiotic-based vaccine, L3-SARS, by integrating a gene fragment that encodes the spike protein S of the SARS-CoV-2 virus into the genome of the probiotic strain E. faecium L3, inducing the expression of viral antigen on the surface of bacteria. Previous studies demonstrated the efficacy of this vaccine candidate in providing protection against the virus in Syrian hamsters. In this present study, utilizing laboratory mice, we assess the immune response subsequent to immunization via the gastrointestinal mucosa and discuss its potential as an initial phase in a two-stage vaccination strategy. Our findings indicate that the oral administration of L3-SARS elicits an adaptive immune response in mice. Pre-immunization with L3-SARS enhances and prolongs the humoral immune response following a single subcutaneous immunization with a recombinant S-protein analogous to the S-insert of the coronavirus in Enterococcus faecium L3.
Collapse
Affiliation(s)
| | | | - Yulia Desheva
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the World-Class Research Center, Center for Personalized Medicine, FSBSI, IEM, 197376 Saint Petersburg, Russia; (G.L.); (T.G.); (T.K.); (E.B.); (I.K.); (O.K.); (A.S.)
| | | | | | | | | | | |
Collapse
|
6
|
Raadsen MP, Visser C, Lavell AHA, van de Munckhof AAGA, Coutinho JM, de Maat MPM, GeurtsvanKessel CH, Amsterdam UMC COVID-19 S3/HCW Study Group, Bomers MK, Haagmans BL, van Gorp ECM, Porcelijn L, Kruip MJHA. Transient Autoreactive PF4 and Antiphospholipid Antibodies in COVID-19 Vaccine Recipients. Vaccines (Basel) 2023; 11:1851. [PMID: 38140254 PMCID: PMC10747426 DOI: 10.3390/vaccines11121851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Vaccine-induced immune thrombotic thrombocytopenia (VITT) is a rare autoimmune condition associated with recombinant adenovirus (rAV)-based COVID-19 vaccines. It is thought to arise from autoantibodies targeting platelet factor 4 (aPF4), triggered by vaccine-induced inflammation and the formation of neo-antigenic complexes between PF4 and the rAV vector. To investigate the specific induction of aPF4 by rAV-based vaccines, we examined sera from rAV vaccine recipients (AZD1222, AD26.COV2.S) and messenger RNA (mRNA) based (mRNA-1273, BNT162b2) COVID-19 vaccine recipients. We compared the antibody fold change (FC) for aPF4 and for antiphospholipid antibodies (aPL) of rAV to mRNA vaccine recipients. We combined two biobanks of Dutch healthcare workers and matched rAV-vaccinated individuals to mRNA-vaccinated controls, based on age, sex and prior history of COVID-19 (AZD1222: 37, Ad26.COV2.S: 35, mRNA-1273: 47, BNT162b2: 26). We found no significant differences in aPF4 FCs after the first (0.99 vs. 1.08, mean difference (MD) = -0.11 (95% CI -0.23 to 0.057)) and second doses of AZD1222 (0.99 vs. 1.10, MD = -0.11 (95% CI -0.31 to 0.10)) and after a single dose of Ad26.COV2.S compared to mRNA-based vaccines (1.01 vs. 0.99, MD = 0.026 (95% CI -0.13 to 0.18)). The mean FCs for the aPL in rAV-based vaccine recipients were similar to those in mRNA-based vaccines. No correlation was observed between post-vaccination aPF4 levels and vaccine type (mean aPF difference -0.070 (95% CI -0.14 to 0.002) mRNA vs. rAV). In summary, our study indicates that rAV and mRNA-based COVID-19 vaccines do not substantially elevate aPF4 levels in healthy individuals.
Collapse
Affiliation(s)
- Matthijs P. Raadsen
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (M.P.R.); (C.H.G.); (B.L.H.); (E.C.M.v.G.)
| | - Chantal Visser
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (C.V.); (M.P.M.d.M.)
| | - A. H. Ayesha Lavell
- Department of Internal Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.H.A.L.); (M.K.B.)
- Amsterdam Institute for Infection & Immunity, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Anita A. G. A. van de Munckhof
- Department of Neurology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.A.G.A.v.d.M.); (J.M.C.)
| | - Jonathan M. Coutinho
- Department of Neurology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.A.G.A.v.d.M.); (J.M.C.)
| | - Moniek P. M. de Maat
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (C.V.); (M.P.M.d.M.)
| | - Corine H. GeurtsvanKessel
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (M.P.R.); (C.H.G.); (B.L.H.); (E.C.M.v.G.)
| | | | - Marije K. Bomers
- Department of Internal Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.H.A.L.); (M.K.B.)
- Amsterdam Institute for Infection & Immunity, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Bart L. Haagmans
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (M.P.R.); (C.H.G.); (B.L.H.); (E.C.M.v.G.)
| | - Eric C. M. van Gorp
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (M.P.R.); (C.H.G.); (B.L.H.); (E.C.M.v.G.)
| | - Leendert Porcelijn
- Department of Immunohematology Diagnostics, Sanquin Diagnostic Services, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands;
| | - Marieke J. H. A. Kruip
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (C.V.); (M.P.M.d.M.)
| |
Collapse
|
7
|
Martin TM, Robinson ST, Huang Y. Discovery medicine - the HVTN's iterative approach to developing an HIV-1 broadly neutralizing vaccine. Curr Opin HIV AIDS 2023; 18:290-299. [PMID: 37712873 PMCID: PMC10552837 DOI: 10.1097/coh.0000000000000821] [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] [Indexed: 09/16/2023]
Abstract
PURPOSE OF REVIEW In the past two decades, there has been an explosion in the discovery of HIV-1 broadly neutralizing antibodies (bnAbs) and associated vaccine strategies to induce them. This abundance of approaches necessitates a system that accurately and expeditiously identifies the most promising regimens. We herein briefly review the background science of bnAbs, provide a description of the first round of phase 1 discovery medicine studies, and suggest an approach to integrate these into a comprehensive HIV-1-neutralizing vaccine. RECENT FINDINGS With recent preclinical success including induction of early stage bnAbs in mouse knockin models and rhesus macaques, successful priming of VRC01-class bnAbs with eOD-GT8 in a recent study in humans, and proof-of-concept that intravenous infusion of VRC01 prevents sexual transmission of virus in humans, the stage is set for a broad and comprehensive bnAb vaccine program. Leveraging significant advances in protein nanoparticle science, mRNA technology, adjuvant development, and B-cell and antibody analyses, the HVTN has reconfigured its HIV-1 vaccine strategy by developing the Discovery Medicine Program to test promising vaccine candidates targeting six key epitopes. SUMMARY The HVTN Discovery Medicine program is testing multiple HIV-1-neutralizing vaccine candidates.
Collapse
Affiliation(s)
- Troy M Martin
- Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | | |
Collapse
|
8
|
Planchais C, Molinos-Albert LM, Rosenbaum P, Hieu T, Kanyavuz A, Clermont D, Prazuck T, Lefrou L, Dimitrov JD, Hüe S, Hocqueloux L, Mouquet H. HIV-1 treatment timing shapes the human intestinal memory B-cell repertoire to commensal bacteria. Nat Commun 2023; 14:6326. [PMID: 37816704 PMCID: PMC10564866 DOI: 10.1038/s41467-023-42027-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
HIV-1 infection causes severe alterations of gut mucosa, microbiota and immune system, which can be curbed by early antiretroviral therapy. Here, we investigate how treatment timing affects intestinal memory B-cell and plasmablast repertoires of HIV-1-infected humans. We show that only class-switched memory B cells markedly differ between subjects treated during the acute and chronic phases of infection. Intestinal memory B-cell monoclonal antibodies show more prevalent polyreactive and commensal bacteria-reactive clones in late- compared to early-treated individuals. Mirroring this, serum IgA polyreactivity and commensal-reactivity are strongly increased in late-treated individuals and correlate with intestinal permeability and systemic inflammatory markers. Polyreactive blood IgA memory B cells, many of which egressed from the gut, are also substantially enriched in late-treated individuals. Our data establish gut and systemic B-cell polyreactivity to commensal bacteria as hallmarks of chronic HIV-1 infection and suggest that initiating treatment early may limit intestinal B-cell abnormalities compromising HIV-1 humoral response.
Collapse
Affiliation(s)
- Cyril Planchais
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cité, INSERM U1222, F-75015, Paris, France
| | - Luis M Molinos-Albert
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cité, INSERM U1222, F-75015, Paris, France
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036, Barcelona, Spain
| | - Pierre Rosenbaum
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cité, INSERM U1222, F-75015, Paris, France
| | - Thierry Hieu
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cité, INSERM U1222, F-75015, Paris, France
| | - Alexia Kanyavuz
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006, Paris, France
| | - Dominique Clermont
- Collection of the Institut Pasteur, Institut Pasteur, Université Paris Cité, 75015, Paris, France
| | - Thierry Prazuck
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, 45067, Orléans, France
| | - Laurent Lefrou
- Service d'Hépato-Gastro-Entérologie, CHR d'Orléans-La Source, 45067, Orléans, France
| | - Jordan D Dimitrov
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006, Paris, France
| | - Sophie Hüe
- INSERM U955-Équipe 16, Université Paris-Est Créteil, Faculté de Médecine, 94000, Créteil, France
| | - Laurent Hocqueloux
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, 45067, Orléans, France
| | - Hugo Mouquet
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cité, INSERM U1222, F-75015, Paris, France.
| |
Collapse
|
9
|
Autoantibodies against chemokines post-SARS-CoV-2 infection correlate with disease course. Nat Immunol 2023; 24:604-611. [PMID: 36879067 PMCID: PMC10063443 DOI: 10.1038/s41590-023-01445-w] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/27/2023] [Indexed: 03/08/2023]
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 associates with diverse symptoms, which can persist for months. While antiviral antibodies are protective, those targeting interferons and other immune factors are associated with adverse coronavirus disease 2019 (COVID-19) outcomes. Here we discovered that antibodies against specific chemokines were omnipresent post-COVID-19, were associated with favorable disease outcome and negatively correlated with the development of long COVID at 1 yr post-infection. Chemokine antibodies were also present in HIV-1 infection and autoimmune disorders, but they targeted different chemokines compared with COVID-19. Monoclonal antibodies derived from COVID-19 convalescents that bound to the chemokine N-loop impaired cell migration. Given the role of chemokines in orchestrating immune cell trafficking, naturally arising chemokine antibodies may modulate the inflammatory response and thus bear therapeutic potential.
Collapse
|
10
|
Haynes BF, Wiehe K, Borrow P, Saunders KO, Korber B, Wagh K, McMichael AJ, Kelsoe G, Hahn BH, Alt F, Shaw GM. Strategies for HIV-1 vaccines that induce broadly neutralizing antibodies. Nat Rev Immunol 2023; 23:142-158. [PMID: 35962033 PMCID: PMC9372928 DOI: 10.1038/s41577-022-00753-w] [Citation(s) in RCA: 180] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2022] [Indexed: 01/07/2023]
Abstract
After nearly four decades of research, a safe and effective HIV-1 vaccine remains elusive. There are many reasons why the development of a potent and durable HIV-1 vaccine is challenging, including the extraordinary genetic diversity of HIV-1 and its complex mechanisms of immune evasion. HIV-1 envelope glycoproteins are poorly recognized by the immune system, which means that potent broadly neutralizing antibodies (bnAbs) are only infrequently induced in the setting of HIV-1 infection or through vaccination. Thus, the biology of HIV-1-host interactions necessitates novel strategies for vaccine development to be designed to activate and expand rare bnAb-producing B cell lineages and to select for the acquisition of critical improbable bnAb mutations. Here we discuss strategies for the induction of potent and broad HIV-1 bnAbs and outline the steps that may be necessary for ultimate success.
Collapse
Affiliation(s)
- Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Department of Immunology, Duke University of School of Medicine, Durham, NC, USA.
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Persephone Borrow
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Bette Korber
- T-6: Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
- New Mexico Consortium, Los Alamos, NM, USA
| | - Kshitij Wagh
- T-6: Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
- New Mexico Consortium, Los Alamos, NM, USA
| | - Andrew J McMichael
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Garnett Kelsoe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Immunology, Duke University of School of Medicine, Durham, NC, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Beatrice H Hahn
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Frederick Alt
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Department of Genetics, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA
| | - George M Shaw
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Pilewski KA, Wall S, Richardson SI, Manamela NP, Clark K, Hermanus T, Binshtein E, Venkat R, Sautto GA, Kramer KJ, Shiakolas AR, Setliff I, Salas J, Mapengo RE, Suryadevara N, Brannon JR, Beebout CJ, Parks R, Raju N, Frumento N, Walker LM, Fechter EF, Qin JS, Murji AA, Janowska K, Thakur B, Lindenberger J, May AJ, Huang X, Sammour S, Acharya P, Carnahan RH, Ross TM, Haynes BF, Hadjifrangiskou M, Crowe JE, Bailey JR, Kalams S, Morris L, Georgiev IS. Functional HIV-1/HCV cross-reactive antibodies isolated from a chronically co-infected donor. Cell Rep 2023; 42:112044. [PMID: 36708513 PMCID: PMC10372200 DOI: 10.1016/j.celrep.2023.112044] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/30/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Despite prolific efforts to characterize the antibody response to human immunodeficiency virus type 1 (HIV-1) and hepatitis C virus (HCV) mono-infections, the response to chronic co-infection with these two ever-evolving viruses is poorly understood. Here, we investigate the antibody repertoire of a chronically HIV-1/HCV co-infected individual using linking B cell receptor to antigen specificity through sequencing (LIBRA-seq). We identify five HIV-1/HCV cross-reactive antibodies demonstrating binding and functional cross-reactivity between HIV-1 and HCV envelope glycoproteins. All five antibodies show exceptional HCV neutralization breadth and effector functions against both HIV-1 and HCV. One antibody, mAb688, also cross-reacts with influenza and coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We examine the development of these antibodies using next-generation sequencing analysis and lineage tracing and find that somatic hypermutation established and enhanced this reactivity. These antibodies provide a potential future direction for therapeutic and vaccine development against current and emerging infectious diseases. More broadly, chronic co-infection represents a complex immunological challenge that can provide insights into the fundamental rules that underly antibody-antigen specificity.
Collapse
Affiliation(s)
- Kelsey A Pilewski
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Steven Wall
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Simone I Richardson
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Nelia P Manamela
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa
| | - Kaitlyn Clark
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tandile Hermanus
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa
| | - Elad Binshtein
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rohit Venkat
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Giuseppe A Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
| | - Kevin J Kramer
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Andrea R Shiakolas
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ian Setliff
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jordan Salas
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rutendo E Mapengo
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa
| | - Naveen Suryadevara
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John R Brannon
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Connor J Beebout
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rob Parks
- Duke Human Vaccine Institute, Duke University, Durham, NC 27710, USA
| | - Nagarajan Raju
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nicole Frumento
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lauren M Walker
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Juliana S Qin
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Amyn A Murji
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Bhishem Thakur
- Duke Human Vaccine Institute, Duke University, Durham, NC 27710, USA
| | | | - Aaron J May
- Duke Human Vaccine Institute, Duke University, Durham, NC 27710, USA
| | - Xiao Huang
- Duke Human Vaccine Institute, Duke University, Durham, NC 27710, USA
| | - Salam Sammour
- Duke Human Vaccine Institute, Duke University, Durham, NC 27710, USA
| | - Priyamvada Acharya
- Duke Human Vaccine Institute, Duke University, Durham, NC 27710, USA; Department of Biochemistry, Duke University, Durham, NC 27710, USA; Department of Surgery, Duke University, Durham, NC 27710, USA
| | - Robert H Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Barton F Haynes
- Departments of Medicine and Immunology, Duke University, Durham, NC 27710, USA; Duke Human Vaccine Institute, Duke University, Durham, NC 27710, USA
| | - Maria Hadjifrangiskou
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Justin R Bailey
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Spyros Kalams
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lynn Morris
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Ivelin S Georgiev
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Computer Science, Vanderbilt University, Nashville, TN 37232, USA; Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA; Program in Computational Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
12
|
Mallaby J, Ng J, Stewart A, Sinclair E, Dunn-Walters D, Hershberg U. Chickens, more than humans, focus the diversity of their immunoglobulin genes on the complementarity-determining region but utilise amino acids, indicative of a more cross-reactive antibody repertoire. Front Immunol 2022; 13:837246. [PMID: 36569888 PMCID: PMC9772431 DOI: 10.3389/fimmu.2022.837246] [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: 12/16/2021] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
The mechanisms of B-cell diversification differ greatly between aves and mammals, but both produce B cells and antibodies capable of supporting an effective immune response. To see how differences in the generation of diversity might affect overall repertoire diversity, we have compared the diversity characteristics of immunoglobulin genes from domestic chickens to those from humans. Both use V(D)J gene rearrangement and somatic hypermutation, but only chickens use somatic gene conversion. A range of diversity analysis tools were used to investigate multiple aspects of amino acid diversity at both the germline and repertoire levels. The effect of differing amino acid usages on antibody characteristics was assessed. At both the germline and repertoire levels, chickens exhibited lower amino acid diversity in comparison to the human immunoglobulin genes, especially outside of the complementarity-determining region (CDR). Chickens were also found to possess much larger and more hydrophilic CDR3s with a higher predicted protein binding potential, suggesting that the antigen-binding site in chicken antibodies is more flexible and more polyreactive than that seen in human antibodies.
Collapse
Affiliation(s)
- Jessica Mallaby
- Department of Bioscience and Medicine, University of Surrey, Guildford, United Kingdom
| | - Joseph Ng
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, United Kingdom
| | - Alex Stewart
- Department of Bioscience and Medicine, University of Surrey, Guildford, United Kingdom
| | - Emma Sinclair
- Department of Bioscience and Medicine, University of Surrey, Guildford, United Kingdom
| | - Deborah Dunn-Walters
- Department of Bioscience and Medicine, University of Surrey, Guildford, United Kingdom
| | - Uri Hershberg
- Department of Human Biology, University of Haifa, Haifa, Israel
| |
Collapse
|
13
|
Jaiswal D, Verma S, Nair DT, Salunke DM. Antibody multispecificity: A necessary evil? Mol Immunol 2022; 152:153-161. [DOI: 10.1016/j.molimm.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022]
|
14
|
Muri J, Cecchinato V, Cavalli A, Shanbhag AA, Matkovic M, Biggiogero M, Maida PA, Moritz J, Toscano C, Ghovehoud E, Furlan R, Barbic F, Voza A, Nadai GD, Cervia C, Zurbuchen Y, Taeschler P, Murray LA, Danelon-Sargenti G, Moro S, Gong T, Piffaretti P, Bianchini F, Crivelli V, Podešvová L, Pedotti M, Jarrossay D, Sgrignani J, Thelen S, Uhr M, Bernasconi E, Rauch A, Manzo A, Ciurea A, Rocchi MB, Varani L, Moser B, Bottazzi B, Thelen M, Fallon BA, Boyman O, Mantovani A, Garzoni C, Franzetti-Pellanda A, Uguccioni M, Robbiani DF. Anti-chemokine antibodies after SARS-CoV-2 infection correlate with favorable disease course. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.05.23.493121. [PMID: 35664993 PMCID: PMC9164443 DOI: 10.1101/2022.05.23.493121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Infection by SARS-CoV-2 leads to diverse symptoms, which can persist for months. While antiviral antibodies are protective, those targeting interferons and other immune factors are associated with adverse COVID-19 outcomes. Instead, we discovered that antibodies against specific chemokines are omnipresent after COVID-19, associated with favorable disease, and predictive of lack of long COVID symptoms at one year post infection. Anti-chemokine antibodies are present also in HIV-1 infection and autoimmune disorders, but they target different chemokines than those in COVID-19. Monoclonal antibodies derived from COVID- 19 convalescents that bind to the chemokine N-loop impair cell migration. Given the role of chemokines in orchestrating immune cell trafficking, naturally arising anti-chemokine antibodies associated with favorable COVID-19 may be beneficial by modulating the inflammatory response and thus bear therapeutic potential. One-Sentence Summary Naturally arising anti-chemokine antibodies associate with favorable COVID-19 and predict lack of long COVID.
Collapse
Affiliation(s)
- Jonathan Muri
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Valentina Cecchinato
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
- Swiss Institute of Bioinformatics; Lausanne, Switzerland
| | - Akanksha A. Shanbhag
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Milos Matkovic
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Maira Biggiogero
- Clinical Research Unit, Clinica Luganese Moncucco; Lugano, Switzerland
| | - Pier Andrea Maida
- Clinical Research Unit, Clinica Luganese Moncucco; Lugano, Switzerland
| | - Jacques Moritz
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Chiara Toscano
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Elaheh Ghovehoud
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Raffaello Furlan
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
- Internal Medicine, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Franca Barbic
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
- Internal Medicine, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Antonio Voza
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
- Department of Emergency, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Guendalina De Nadai
- Emergency Medicine Residency School, Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4 - 20090 Pieve Emanuele, Milan, Italy
| | - Carlo Cervia
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Yves Zurbuchen
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Patrick Taeschler
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lilly A. Murray
- Lyme & Tick-Borne Diseases Research Center at Columbia University Irving Medical Center, New York, NY, USA
| | | | - Simone Moro
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Tao Gong
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Pietro Piffaretti
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Filippo Bianchini
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Virginia Crivelli
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Lucie Podešvová
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Mattia Pedotti
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - David Jarrossay
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Jacopo Sgrignani
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Sylvia Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | | | - Enos Bernasconi
- Regional Hospital Lugano, Ente Ospedaliero Cantonale; Lugano, Switzerland
- Università della Svizzera italiana; Lugano, Switzerland
| | - Andri Rauch
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern; Bern, Switzerland
| | - Antonio Manzo
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Division of Rheumatology, IRCCS Policlinico San Matteo Foundation, University of Pavia; Pavia, Italy
| | - Adrian Ciurea
- Department of Rheumatology, Zurich University Hospital, University of Zurich; Zurich, Switzerland
| | - Marco B.L. Rocchi
- Department of Biomolecular Sciences, Biostatistics Unit, University of Urbino; Urbino, Italy
| | - Luca Varani
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Bernhard Moser
- Division of Infection & Immunity, Henry Wellcome Building, Cardiff University School of Medicine; Cardiff, United Kingdom
| | - Barbara Bottazzi
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| | - Brian A. Fallon
- Lyme & Tick-Borne Diseases Research Center at Columbia University Irving Medical Center, New York, NY, USA
- Lyme Research Program at the New York State Psychiatric Institute, New York, NY, USA
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Faculty of Medicine and Faculty of Science, University of Zurich, Zurich, Switzerland
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - Christian Garzoni
- Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco; Lugano, Switzerland
| | | | - Mariagrazia Uguccioni
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Davide F. Robbiani
- Institute for Research in Biomedicine, Università della Svizzera italiana; Bellinzona, Switzerland
| |
Collapse
|
15
|
Li X, Liao D, Li Z, Li J, Diaz M, Verkoczy L, Gao F. Autoreactivity and broad neutralization of antibodies against HIV-1 are governed by distinct mutations: Implications for vaccine design strategies. Front Immunol 2022; 13:977630. [PMID: 36479128 PMCID: PMC9720396 DOI: 10.3389/fimmu.2022.977630] [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: 06/24/2022] [Accepted: 11/04/2022] [Indexed: 11/22/2022] Open
Abstract
Many of the best HIV-1 broadly neutralizing antibodies (bnAbs) known have poly-/autoreactive features that disfavor normal B cell development and maturation, posing a major hurdle in developing an effective HIV-1 vaccine. Key to resolving this problem is to understand if, and to what extent, neutralization breadth-conferring mutations acquired by bnAbs contribute to their autoreactivity. Here, we back-mutated all known changes made by a prototype CD4 binding site-directed bnAb lineage, CH103-106, during its later maturation steps. Strikingly, of 29 mutations examined, only four were crucial for increased autoreactivity, with minimal or no impact on neutralization. Furthermore, three of these residues were clustered in the heavy chain complementarity-determining region 2 (HCDR2). Our results demonstrate that broad neutralization activity and autoreactivity in the CH103-106 bnAb lineage can be governed by a few, distinct mutations during maturation. This provides strong rationale for developing immunogens that favor bnAb lineages bearing "neutralization-only" mutations into current HIV-1 vaccine designs.
Collapse
Affiliation(s)
- Xiaojun Li
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Dongmei Liao
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| | - Zhengyang Li
- School of Life Sciences, Fudan University, Shanghai, China
| | - Jixi Li
- School of Life Sciences, Fudan University, Shanghai, China
| | - Marilyn Diaz
- Applied Biomedical Science Institute, San Diego, CA, United States
| | - Laurent Verkoczy
- Applied Biomedical Science Institute, San Diego, CA, United States
| | - Feng Gao
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Institute of Molecular and Medical Virology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, Guangdongg, China
| |
Collapse
|
16
|
Ausserwöger H, Schneider MM, Herling TW, Arosio P, Invernizzi G, Knowles TPJ, Lorenzen N. Non-specificity as the sticky problem in therapeutic antibody development. Nat Rev Chem 2022; 6:844-861. [PMID: 37117703 DOI: 10.1038/s41570-022-00438-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2022] [Indexed: 11/16/2022]
Abstract
Antibodies are highly potent therapeutic scaffolds with more than a hundred different products approved on the market. Successful development of antibody-based drugs requires a trade-off between high target specificity and target binding affinity. In order to better understand this problem, we here review non-specific interactions and explore their fundamental physicochemical origins. We discuss the role of surface patches - clusters of surface-exposed amino acid residues with similar physicochemical properties - as inducers of non-specific interactions. These patches collectively drive interactions including dipole-dipole, π-stacking and hydrophobic interactions to complementary moieties. We elucidate links between these supramolecular assembly processes and macroscopic development issues, such as decreased physical stability and poor in vivo half-life. Finally, we highlight challenges and opportunities for optimizing protein binding specificity and minimizing non-specificity for future generations of therapeutics.
Collapse
|
17
|
Haruna S, Takeda K, El-Hussien MA, Maeda Y, Hayama M, Shikina T, Doi K, Inohara H, Kikutani H, Sakakibara S. Local production of broadly cross-reactive IgE against multiple fungal cell wall polysaccharides in patients with allergic fungal rhinosinusitis. Allergy 2022; 77:3147-3151. [PMID: 35716354 DOI: 10.1111/all.15413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/06/2022] [Accepted: 06/15/2022] [Indexed: 01/28/2023]
Affiliation(s)
- Soichiro Haruna
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Kazuya Takeda
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Otolaryngology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Marwa Ali El-Hussien
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yohei Maeda
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masaki Hayama
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | - Katsumi Doi
- Department of Otolaryngology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hitoshi Kikutani
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shuhei Sakakibara
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
18
|
França RKADO, Silva JM, Rodrigues LS, Sokolowskei D, Brigido MM, Maranhão AQ. New Anti-Flavivirus Fusion Loop Human Antibodies with Zika Virus-Neutralizing Potential. Int J Mol Sci 2022; 23:ijms23147805. [PMID: 35887153 PMCID: PMC9321016 DOI: 10.3390/ijms23147805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 02/04/2023] Open
Abstract
Zika virus infections exhibit recurrent outbreaks and can be responsible for disease complications such as congenital Zika virus syndrome. Effective therapeutic interventions are still a challenge. Antibodies can provide significant protection, although the antibody response may fail due to antibody-dependent enhancement reactions. The choice of the target antigen is a crucial part of the process to generate effective neutralizing antibodies. Human anti-Zika virus antibodies were selected by phage display technology. The antibodies were selected against a mimetic peptide based on the fusion loop region in the protein E of Zika virus, which is highly conserved among different flaviviruses. Four rounds of selection were performed using the synthetic peptide in two strategies: the first was using the acidic elution of bound phages, and the second was by applying a competing procedure. After panning, the selected VH and VL domains were determined by combining NGS and bioinformatic approaches. Three different human monoclonal antibodies were expressed as scFvs and further characterized. All showed a binding capacity to Zika (ZIKV) and showed cross-recognition with yellow fever (YFV) and dengue (DENV) viruses. Two of these antibodies, AZ1p and AZ6m, could neutralize the ZIKV infection in vitro. Due to the conservation of the fusion loop region, these new antibodies can potentially be used in therapeutic intervention against Zika virus and other flavivirus illnesses.
Collapse
Affiliation(s)
- Renato Kaylan Alves de Oliveira França
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
- Graduation Program in Molecular Pathology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Jacyelle Medeiros Silva
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
| | - Lucas Silva Rodrigues
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
- Graduation Program in Molecular Pathology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Dimitri Sokolowskei
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
- Graduation Program in Molecular Biology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Marcelo Macedo Brigido
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
- Graduation Program in Molecular Pathology, University of Brasilia, Brasilia 70910-900, Brazil
- Graduation Program in Molecular Biology, University of Brasilia, Brasilia 70910-900, Brazil
- III-Immunology Investigation Institute–CNPq-MCT, São Paulo 05403-000, Brazil
- Correspondence:
| | - Andrea Queiroz Maranhão
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
- Graduation Program in Molecular Pathology, University of Brasilia, Brasilia 70910-900, Brazil
- Graduation Program in Molecular Biology, University of Brasilia, Brasilia 70910-900, Brazil
- III-Immunology Investigation Institute–CNPq-MCT, São Paulo 05403-000, Brazil
| |
Collapse
|
19
|
Bonduelle O, Chaudesaigues C, Tolazzi M, Suleiman E, de Bernard S, Alves K, Nourikyan J, Bohec M, Baudrin LG, Katinger D, Debré P, Scarlatti G, Vieillard V, Combadière B. Dichotomy in Neutralizing Antibody Induction to Peptide-Conjugated Vaccine in Squalene Emulsion Contrast With Aluminum Hydroxide Formulation. Front Immunol 2022; 13:848571. [PMID: 35464449 PMCID: PMC9021396 DOI: 10.3389/fimmu.2022.848571] [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: 01/04/2022] [Accepted: 02/22/2022] [Indexed: 01/28/2023] Open
Abstract
W614A-3S peptide is a modified 3S motif of the HIV-gp41 (mutation W614A). We previously detected the presence of natural neutralizing antibodies directed against W614A-3S peptide (NAbs) in long-term non-progressor HIV+ patients. Here, we compared the efficacy of W614A-3S peptide formulated in either squalene emulsion (SQE) or in aluminum hydroxide (Alum) in inducing broadly-NAbs (bNAbs). Rabbit and mouse models were used to screen the induction of bNAbs following 4 immunizations. SQE was more efficient than Alum formulation in inducing W614A-3S-specific bNAbs with up to 67%-93% of HIV strains neutralized. We then analyzed the quality of peptide-specific murine B cells by single-cell gene expression by quantitative reverse transcription-PCR and single-cell V(D)J sequencing. We found more frequent germinal center B cells in SQE than in Alum, albeit with a different gene expression profile. The V(D)J sequencing of W614A-3S-specific BCR showed significant differences in BCR sequences and validates the dichotomy between adjuvant formulations. All sixteen BCR sequences which were cloned were specific of peptide. Adjuvant formulations of W614A-3S-peptide-conjugated immunogen impact the quantity and quality of B cell immune responses at both the gene expression level and BCR sequence.
Collapse
Affiliation(s)
- Olivia Bonduelle
- Sorbonne Université, Institut national de la santé et de la recherche médicale (Inserm) U1135, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Chloé Chaudesaigues
- Sorbonne Université, Institut national de la santé et de la recherche médicale (Inserm) U1135, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Monica Tolazzi
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Ehsan Suleiman
- Polymun Scientific Immunbiologische Forschung GmbH, Klosterneuburg, Austria
| | | | | | | | - Mylene Bohec
- Institut Curie, Genomics of Excellence (ICGex) Platform, Paris Science et Lettres (PSL) Research University, Paris, France
| | - Laura G Baudrin
- Institut Curie, Genomics of Excellence (ICGex) Platform, Paris Science et Lettres (PSL) Research University, Paris, France
| | - Dietmar Katinger
- Polymun Scientific Immunbiologische Forschung GmbH, Klosterneuburg, Austria
| | - Patrice Debré
- Sorbonne Université, Institut national de la santé et de la recherche médicale (Inserm) U1135, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Vincent Vieillard
- Sorbonne Université, Institut national de la santé et de la recherche médicale (Inserm) U1135, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Behazine Combadière
- Sorbonne Université, Institut national de la santé et de la recherche médicale (Inserm) U1135, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| |
Collapse
|
20
|
Parallelism of intestinal secretory IgA shapes functional microbial fitness. Nature 2021; 598:657-661. [PMID: 34646015 DOI: 10.1038/s41586-021-03973-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 08/31/2021] [Indexed: 12/19/2022]
Abstract
Dimeric IgA secreted across mucous membranes in response to nonpathogenic taxa of the microbiota accounts for most antibody production in mammals. Diverse binding specificities can be detected within the polyclonal mucosal IgA antibody response1-10, but limited monoclonal hybridomas have been studied to relate antigen specificity or polyreactive binding to functional effects on microbial physiology in vivo11-17. Here we use recombinant dimeric monoclonal IgAs (mIgAs) to finely map the intestinal plasma cell response to microbial colonization with a single microorganism in mice. We identify a range of antigen-specific mIgA molecules targeting defined surface and nonsurface membrane antigens. Secretion of individual dimeric mIgAs targeting different antigens in vivo showed distinct alterations in the function and metabolism of intestinal bacteria, largely through specific binding. Even in cases in which the same microbial antigen is targeted, microbial metabolic alterations differed depending on IgA epitope specificity. By contrast, bacterial surface coating generally reduced motility and limited bile acid toxicity. The overall intestinal IgA response to a single microbe therefore contains parallel components with distinct effects on microbial carbon-source uptake, bacteriophage susceptibility, motility and membrane integrity.
Collapse
|
21
|
Li L, Dugan HL, Stamper CT, Lan LYL, Asby NW, Knight M, Stovicek O, Zheng NY, Madariaga ML, Shanmugarajah K, Jansen MO, Changrob S, Utset HA, Henry C, Nelson C, Jedrzejczak RP, Fremont DH, Joachimiak A, Krammer F, Huang J, Khan AA, Wilson PC. Improved integration of single-cell transcriptome and surface protein expression by LinQ-View. CELL REPORTS METHODS 2021; 1:100056. [PMID: 35475142 PMCID: PMC9017149 DOI: 10.1016/j.crmeth.2021.100056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/12/2021] [Accepted: 06/25/2021] [Indexed: 12/26/2022]
Abstract
Multimodal advances in single-cell sequencing have enabled the simultaneous quantification of cell surface protein expression alongside unbiased transcriptional profiling. Here, we present LinQ-View, a toolkit designed for multimodal single-cell data visualization and analysis. LinQ-View integrates transcriptional and cell surface protein expression profiling data to reveal more accurate cell heterogeneity and proposes a quantitative metric for cluster purity assessment. Through comparison with existing multimodal methods on multiple public CITE-seq datasets, we demonstrate that LinQ-View efficiently generates accurate cell clusters, especially in CITE-seq data with routine numbers of surface protein features, by preventing variations in a single surface protein feature from affecting results. Finally, we utilized this method to integrate single-cell transcriptional and protein expression data from SARS-CoV-2-infected patients, revealing antigen-specific B cell subsets after infection. Our results suggest LinQ-View could be helpful for multimodal analysis and purity assessment of CITE-seq datasets that target specific cell populations (e.g., B cells).
Collapse
Affiliation(s)
- Lei Li
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Haley L. Dugan
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | | | - Linda Yu-Ling Lan
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Nicholas W. Asby
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Matthew Knight
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Olivia Stovicek
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Nai-Ying Zheng
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | | | | | - Maud O. Jansen
- Section of Hospital Medicine, University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Siriruk Changrob
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Henry A. Utset
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Carole Henry
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Christopher Nelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robert P. Jedrzejczak
- Center for Structural Genomics of Infectious Diseases, Consortium for Advanced Science and Engineering, University of Chicago, Chicago, IL 60637, USA
- Structural Biology Center, X-ray Science Division, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Daved H. Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrzej Joachimiak
- Center for Structural Genomics of Infectious Diseases, Consortium for Advanced Science and Engineering, University of Chicago, Chicago, IL 60637, USA
- Structural Biology Center, X-ray Science Division, Argonne National Laboratory, Lemont, IL 60439, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jun Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Aly A. Khan
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Patrick C. Wilson
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
22
|
A Germline-Encoded Structural Arginine Trap Underlies the Anti-DNA Reactivity of a Murine V Gene Segment. Int J Mol Sci 2021; 22:ijms22094541. [PMID: 33926148 PMCID: PMC8123574 DOI: 10.3390/ijms22094541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 11/30/2022] Open
Abstract
Autoimmunity may have its origins of early repertoire selection in developmental B cells. Such a primary repertoire is probably shaped by selecting B cells that can efficiently perform productive signaling, stimulated by self-antigens in the bone marrow, such as DNA. In support of that idea, we previously found a V segment from VH10 family that can form antibodies that bind to DNA independent of CDR3 usage. In this paper we designed four antibody fragments in a novel single-chain pre-BCR (scpre-BCR) format containing germinal V gene segments from families known to bind DNA (VH10) or not (VH4) connected to a murine surrogate light chain (SLC), lacking the highly charged unique region (UR), by a hydrophilic peptide linker. We also tested the influence of CDR2 on DNA reactivity by shuffling the CDR2 loop. The scpre-BCRs were expressed in bacteria. VH10 bearing scpre-BCR could bind DNA, while scpre-BCR carrying the VH4 segment did not. The CDR2 loop shuffling hampered VH10 reactivity while displaying a gain-of-function in the nonbinding VH4 germline. We modeled the binding sites demonstrating the conservation of a positivity charged pocket in the VH10 CDR2 as the possible cross-reactive structural element. We presented evidence of DNA reactivity hardwired in a V gene, suggesting a structural mechanism for innate autoreactivity. Therefore, while autoreactivity to DNA can lead to autoimmunity, efficiently signaling for B cell development is likely a trade-off mechanism leading to the selection of potentially autoreactive repertoires.
Collapse
|
23
|
Sáez Moya M, Gutiérrez-Cózar R, Puñet-Ortiz J, Rodríguez de la Concepción ML, Blanco J, Carrillo J, Engel P. Autoimmune B Cell Repertoire in a Mouse Model of Sjögren's Syndrome. Front Immunol 2021; 12:666545. [PMID: 33968069 PMCID: PMC8103202 DOI: 10.3389/fimmu.2021.666545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/07/2021] [Indexed: 11/24/2022] Open
Abstract
In genetically prone individuals, chronic immune activation may lead to expansion of autoreactive lymphocyte clones that can induce organ damage developing autoimmune disorders. Sjögren’s Syndrome (SjS) is a systemic chronic autoimmune disease that primarily affects exocrine glands. Despite the accumulated evidences of profound B-cell alterations of humoral immunity, the repertoire and development of B-cell autoreactivity in SjS remains to be determined. We hypothesize that SjS mice will have an increased frequency of self-reactive B cells with a progressive evolution to antigen-driven oligoclonality. Here, we study the B cell repertoire of NOD.H-2h4 mice, a mouse model of spontaneous autoimmunity mimicking SjS without developing diabetes. A library of 168 hybridomas from NOD.H-2h4 mice and 186 C57BL/6J splenocytes at different ages was created. The presence of mono or polyreactive autoantibodies to several antigens was evaluated by ELISA, and their staining patterns and cellular reactivity were tested by IFA and FACS. We observed a higher frequency of autoreactivity among B-cell clones from NOD.H-2h4 mice as compared to wild-type mice. The presence of polyreactive and autoreactive IgG clones increased with mice age. Strikingly, all anti-Ro52 autoantibodies were polyreactive. No loss of polyreactivity was observed upon antibody class switching to IgG. There was a progression to oligoclonality in IgG B cells with mice aging. Our results indicate that in the NOD.H-2h4 mouse model of SjS, IgG+ B cells are mainly polyreactive and might expand following an unknown antigen-driven positive selection process.
Collapse
Affiliation(s)
- Manuel Sáez Moya
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Rebeca Gutiérrez-Cózar
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Joan Puñet-Ortiz
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | | | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Badalona, Spain, Germans Trias i Pujol Research Institute (IGTP), Catalonia, Spain.,AIDS and Related Diseases Chair, Universitat de Vic-Central de Catalunya (UVIC-UCC), Vic, Spain
| | - Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Badalona, Spain, Germans Trias i Pujol Research Institute (IGTP), Catalonia, Spain
| | - Pablo Engel
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain.,August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| |
Collapse
|
24
|
Demers-Mathieu V, Do DM, Mathijssen GB, Sela DA, Seppo A, Järvinen KM, Medo E. Difference in levels of SARS-CoV-2 S1 and S2 subunits- and nucleocapsid protein-reactive SIgM/IgM, IgG and SIgA/IgA antibodies in human milk. J Perinatol 2021; 41:850-859. [PMID: 32873904 PMCID: PMC7461757 DOI: 10.1038/s41372-020-00805-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/03/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE This study evaluated the presence and the levels of antibodies reactive to SARS-CoV-2 S1 and S2 subunits (S1 + S2), and nucleocapsid protein. STUDY DESIGN The levels of SARS-CoV-2 S1 + S2- and nucleocapsid-reactive SIgM/IgM, IgG and SIgA/IgA were measured in human milk samples from 41 women during the COVID-19 pandemic (2020-HM) and from 16 women 2 years prior to the outbreak (2018-HM). RESULTS SARS-CoV-2 S1 + S2-reactive SIgA/IgA, SIgM/IgM and IgG were detected in 97.6%, 68.3% and 58.5% in human milk whereas nucleocapsid-reactive antibodies were detected in 56.4%, 87.2% and 46.2%, respectively. S1 + S2-reactive IgG was higher in milk from women that had symptoms of viral respiratory infection(s) during the last year than in milk from women without symptom. S1 + S2- and nucleocapsid-reactive IgG were higher in the 2020-HM group compared to the 2018-HM group. CONCLUSIONS The presence of SARS-CoV-2-reactive antibodies in human milk could provide passive immunity to breastfed infants and protect them against COVID-19 diseases.
Collapse
Affiliation(s)
- Veronique Demers-Mathieu
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA.
- Department of Food Science, University of Massachusetts Amherst, Amherst, MA, USA.
| | - Dung M Do
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| | - Gabrielle B Mathijssen
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| | - David A Sela
- Department of Food Science, University of Massachusetts Amherst, Amherst, MA, USA
| | - Antti Seppo
- Department of Pediatrics, Pediatric Allergy/Immunology, University of Rochester, Medical Center, Rochester, NY, USA
| | - Kirsi M Järvinen
- Department of Pediatrics, Pediatric Allergy/Immunology, University of Rochester, Medical Center, Rochester, NY, USA
| | - Elena Medo
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| |
Collapse
|
25
|
The Involvement of Innate and Adaptive Immunity in the Initiation and Perpetuation of Sjögren's Syndrome. Int J Mol Sci 2021; 22:ijms22020658. [PMID: 33440862 PMCID: PMC7826728 DOI: 10.3390/ijms22020658] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/01/2021] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Sjogren’s syndrome (SS) is a chronic autoimmune disease characterized by the infiltration of exocrine glands including salivary and lachrymal glands responsible for the classical dry eyes and mouth symptoms (sicca syndrome). The spectrum of disease manifestations stretches beyond the classical sicca syndrome with systemic manifestations including arthritis, interstitial lung involvement, and neurological involvement. The pathophysiology underlying SS is not well deciphered, but several converging lines of evidence have supported the conjuncture of different factors interplaying together to foster the initiation and perpetuation of the disease. The innate and adaptive immune system play a cardinal role in this process. In this review, we discuss the inherent parts played by both the innate and adaptive immune system in the pathogenesis of SS.
Collapse
|
26
|
Parisis D, Chivasso C, Perret J, Soyfoo MS, Delporte C. Current State of Knowledge on Primary Sjögren's Syndrome, an Autoimmune Exocrinopathy. J Clin Med 2020; 9:E2299. [PMID: 32698400 PMCID: PMC7408693 DOI: 10.3390/jcm9072299] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022] Open
Abstract
Primary Sjögren's syndrome (pSS) is a chronic systemic autoimmune rheumatic disease characterized by lymphoplasmacytic infiltration of the salivary and lacrimal glands, whereby sicca syndrome and/or systemic manifestations are the clinical hallmarks, associated with a particular autoantibody profile. pSS is the most frequent connective tissue disease after rheumatoid arthritis, affecting 0.3-3% of the population. Women are more prone to develop pSS than men, with a sex ratio of 9:1. Considered in the past as innocent collateral passive victims of autoimmunity, the epithelial cells of the salivary glands are now known to play an active role in the pathogenesis of the disease. The aetiology of the "autoimmune epithelitis" still remains unknown, but certainly involves genetic, environmental and hormonal factors. Later during the disease evolution, the subsequent chronic activation of B cells can lead to the development of systemic manifestations or non-Hodgkin's lymphoma. The aim of the present comprehensive review is to provide the current state of knowledge on pSS. The review addresses the clinical manifestations and complications of the disease, the diagnostic workup, the pathogenic mechanisms and the therapeutic approaches.
Collapse
Affiliation(s)
- Dorian Parisis
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium; (D.P.); (C.C.); (J.P.)
- Department of Rheumatology, Erasme Hospital, Université Libre de Bruxelles, 1070 Brussels, Belgium;
| | - Clara Chivasso
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium; (D.P.); (C.C.); (J.P.)
| | - Jason Perret
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium; (D.P.); (C.C.); (J.P.)
| | | | - Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium; (D.P.); (C.C.); (J.P.)
| |
Collapse
|
27
|
Planchais C, Kök A, Kanyavuz A, Lorin V, Bruel T, Guivel-Benhassine F, Rollenske T, Prigent J, Hieu T, Prazuck T, Lefrou L, Wardemann H, Schwartz O, Dimitrov JD, Hocqueloux L, Mouquet H. HIV-1 Envelope Recognition by Polyreactive and Cross-Reactive Intestinal B Cells. Cell Rep 2020; 27:572-585.e7. [PMID: 30970259 PMCID: PMC6458971 DOI: 10.1016/j.celrep.2019.03.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 02/19/2019] [Accepted: 03/08/2019] [Indexed: 12/18/2022] Open
Abstract
Mucosal immune responses to HIV-1 involve the recognition of the viral envelope glycoprotein (gp)160 by tissue-resident B cells and subsequent secretion of antibodies. To characterize the B cells “sensing” HIV-1 in the gut of infected individuals, we probed monoclonal antibodies produced from single intestinal B cells binding to recombinant gp140 trimers. A large fraction of mucosal B cell antibodies were polyreactive and showed only low affinity to HIV-1 envelope glycoproteins, particularly the gp41 moiety. A few high-affinity gp140 antibodies were isolated but lacked neutralizing, potent ADCC, and transcytosis-blocking capacities. Instead, they displayed cross-reactivity with defined self-antigens. Specifically, intestinal HIV-1 gp41 antibodies targeting the heptad repeat 2 region (HR2) cluster II cross-reacted with the p38α mitogen-activated protein kinase 14 (MAPK14). Hence, physiologic polyreactivity of intestinal B cells and molecular mimicry-based self-reactivity of HIV-1 antibodies are two independent phenomena, possibly diverting and/or impairing mucosal humoral immunity to HIV-1. Polyreactive B cells in HIV-1+ intestinal mucosa interact with HIV-1 Env proteins High-affinity intestinal HIV-1 gp140 antibodies display poor antiviral activities Antibodies targeting the gp41 cluster II region cross-react with MAPK14
Collapse
Affiliation(s)
- Cyril Planchais
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Ayrin Kök
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Alexia Kanyavuz
- Sorbonne Universités, UPMC Université Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France
| | - Valérie Lorin
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Timothée Bruel
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Paris 75015, France; CNRS URA3015, Paris, 75015, France
| | - Florence Guivel-Benhassine
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Paris 75015, France; CNRS URA3015, Paris, 75015, France
| | - Tim Rollenske
- Division of B Cell Immunology, German Cancer Research Center, Heidelberg 69120, Germany
| | - Julie Prigent
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Thierry Hieu
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Thierry Prazuck
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, Orléans 45067, France
| | - Laurent Lefrou
- Service d'Hépato-Gastro-Entérologie, CHR d'Orléans-La Source, Orléans 45067, France
| | - Hedda Wardemann
- Division of B Cell Immunology, German Cancer Research Center, Heidelberg 69120, Germany
| | - Olivier Schwartz
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Paris 75015, France; CNRS URA3015, Paris, 75015, France
| | - Jordan D Dimitrov
- Sorbonne Universités, UPMC Université Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France
| | - Laurent Hocqueloux
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, Orléans 45067, France
| | - Hugo Mouquet
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France.
| |
Collapse
|
28
|
Blazkova J, Refsland EW, Clarridge KE, Shi V, Justement JS, Huiting ED, Gittens KR, Chen X, Schmidt SD, Liu C, Doria-Rose N, Mascola JR, Heredia A, Moir S, Chun TW. Glycan-dependent HIV-specific neutralizing antibodies bind to cells of uninfected individuals. J Clin Invest 2020; 129:4832-4837. [PMID: 31589168 DOI: 10.1172/jci125955] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 08/07/2019] [Indexed: 01/09/2023] Open
Abstract
A number of highly potent and broadly neutralizing antibodies (bNAbs) against the human immunodeficiency virus (HIV) have recently been shown to prevent transmission of the virus, suppress viral replication, and delay plasma viral rebound following discontinuation of antiretroviral therapy in animal models and infected humans. However, the degree and extent to which such bNAbs interact with primary lymphocytes have not been fully delineated. Here, we show that certain glycan-dependent bNAbs, such as PGT121 and PGT151, bind to B, activated T, and natural killer (NK) cells of HIV-infected and -uninfected individuals. Binding of these bNAbs, particularly PGT121 and PGT151, to activated CD4+ and CD8+ T cells was mediated by complex-type glycans and was abrogated by enzymatic inhibition of N-linked glycosylation. In addition, a short-term incubation of PGT151 and primary NK cells led to degranulation and cellular death. Our data suggest that the propensity of certain bNAbs to bind uninfected/bystander cells has the potential for unexpected outcomes in passive-transfer studies and underscore the importance of antibody screening against primary lymphocytes.
Collapse
Affiliation(s)
- Jana Blazkova
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID)
| | - Eric W Refsland
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID)
| | - Katherine E Clarridge
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID)
| | - Victoria Shi
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID)
| | - J Shawn Justement
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID)
| | - Erin D Huiting
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID)
| | | | - Xuejun Chen
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland, USA
| | | | - Cuiping Liu
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland, USA
| | | | - John R Mascola
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland, USA
| | - Alonso Heredia
- Institute of Human Virology, University of Maryland, Baltimore, Maryland, USA
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID)
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID)
| |
Collapse
|
29
|
Hua Z, Hou B. The role of B cell antigen presentation in the initiation of CD4+ T cell response. Immunol Rev 2020; 296:24-35. [PMID: 32304104 DOI: 10.1111/imr.12859] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/29/2020] [Accepted: 03/26/2020] [Indexed: 01/21/2023]
Abstract
B cells have been known for their ability to present antigens to T cells for almost 40 years. However, the precise roles of B cell antigen presentation in various immune responses are not completely understood. The term "professional" antigen-presenting cells (APCs) was proposed to distinguish APCs that are required for initiating the immune responses from those use antigen presentation to enhance their own effector functions. Unlike dendritic cells, which are defined as professional APCs for their well-established functions in activating naive T cells, B cells have been shown in the past to mostly present antigens to activated CD4+ T cells mainly to seek help from T helper cells. However, recent evidence suggested that B cells can act as professional APCs under infectious conditions or conditions mimicking viral infections. B cell antigen receptors (BCRs) and the innate receptor Toll-like receptors are activated synergistically in response to pathogens or virus-like particles, under which conditions B cells are not only potent but also the predominant APCs to turn naive CD4+ T cells into T follicular helper cells. The discovery of B cells as professional APCs to initiate CD4+ T cell response provides a new insight for both autoimmune diseases and vaccine development.
Collapse
Affiliation(s)
- Zhaolin Hua
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Baidong Hou
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
30
|
Su B, Dispinseri S, Iannone V, Zhang T, Wu H, Carapito R, Bahram S, Scarlatti G, Moog C. Update on Fc-Mediated Antibody Functions Against HIV-1 Beyond Neutralization. Front Immunol 2019; 10:2968. [PMID: 31921207 PMCID: PMC6930241 DOI: 10.3389/fimmu.2019.02968] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 12/03/2019] [Indexed: 12/31/2022] Open
Abstract
Antibodies (Abs) are the major component of the humoral immune response and a key player in vaccination. The precise Ab-mediated inhibitory mechanisms leading to in vivo protection against HIV have not been elucidated. In addition to the desired viral capture and neutralizing Ab functions, complex Ab-dependent mechanisms that involve engaging immune effector cells to clear infected host cells, immune complexes, and opsonized virus have been proposed as being relevant. These inhibitory mechanisms involve Fc-mediated effector functions leading to Ab-dependent cellular cytotoxicity, phagocytosis, cell-mediated virus inhibition, aggregation, and complement inhibition. Indeed, the decreased risk of infection observed in the RV144 HIV-1 vaccine trial was correlated with the production of non-neutralizing inhibitory Abs, highlighting the role of Ab inhibitory functions besides neutralization. Moreover, Ab isotypes and subclasses recognizing specific HIV envelope epitopes as well as pecular Fc-receptor polymorphisms have been associated with disease progression. These findings further support the need to define which Fc-mediated Ab inhibitory functions leading to protection are critical for HIV vaccine design. Herein, based on our previous review Su & Moog Front Immunol 2014, we update the different inhibitory properties of HIV-specific Abs that may potentially contribute to HIV protection.
Collapse
Affiliation(s)
- Bin Su
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Stefania Dispinseri
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Iannone
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Tong Zhang
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Hao Wu
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Raphael Carapito
- INSERM U1109, LabEx TRANSPLANTEX, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Seiamak Bahram
- INSERM U1109, LabEx TRANSPLANTEX, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Christiane Moog
- INSERM U1109, LabEx TRANSPLANTEX, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Vaccine Research Institute (VRI), Créteil, France
| |
Collapse
|
31
|
Abstract
Adaptive antibody responses provide a crucial means of host defense against viral infections by mediating the neutralization and killing infectious pathogens. At the forefront of humoral defense against viruses lie a subset of innate-like serum antibodies known as natural antibodies (NAbs). NAbs serve multifaceted functions in host defense and play an essential role in early immune responses against viruses. However, there remain many unanswered questions with regard to both the breadth of viral antigens recognized by NAbs, and how B cell ontology and individual antigenic histories intersect to control the development and function of antiviral human NAbs. In the following article we briefly review the current understanding of the functions and source of NAbs in the immune repertoire, their role during antiviral immune responses, the factors influencing the maturation of the NAb repertoire, and finally, the gaps and future research needed to advance our understanding of innate-like B cell biology for the purpose of harnessing NAbs for host defense against viral infections.
Collapse
Affiliation(s)
- J Stewart New
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - R Glenn King
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - John F Kearney
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
32
|
Prigent J, Jarossay A, Planchais C, Eden C, Dufloo J, Kök A, Lorin V, Vratskikh O, Couderc T, Bruel T, Schwartz O, Seaman MS, Ohlenschläger O, Dimitrov JD, Mouquet H. Conformational Plasticity in Broadly Neutralizing HIV-1 Antibodies Triggers Polyreactivity. Cell Rep 2019; 23:2568-2581. [PMID: 29847789 PMCID: PMC5990490 DOI: 10.1016/j.celrep.2018.04.101] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/26/2018] [Accepted: 04/25/2018] [Indexed: 12/18/2022] Open
Abstract
Human high-affinity antibodies to pathogens often recognize unrelated ligands. The molecular origin and the role of this polyreactivity are largely unknown. Here, we report that HIV-1 broadly neutralizing antibodies (bNAbs) are frequently polyreactive, cross-reacting with non-HIV-1 molecules, including self-antigens. Mutating bNAb genes to increase HIV-1 binding and neutralization also results in de novo polyreactivity. Unliganded paratopes of polyreactive bNAbs with improved HIV-1 neutralization exhibit a conformational flexibility, which contributes to enhanced affinity of bNAbs to various HIV-1 envelope glycoproteins and non-HIV antigens. Binding adaptation of polyreactive bNAbs to the divergent ligands mainly involves hydrophophic interactions. Plasticity of bNAbs' paratopes may, therefore, facilitate accommodating divergent viral variants, but it simultaneously triggers promiscuous binding to non-HIV-1 antigens. Thus, a certain level of polyreactivity can be a mark of adaptable antibodies displaying optimal pathogens' recognition.
Collapse
Affiliation(s)
- Julie Prigent
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Annaëlle Jarossay
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France
| | - Cyril Planchais
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Caroline Eden
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jérémy Dufloo
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Paris 75015, France; CNRS URA3015, Paris 75015, France
| | - Ayrin Kök
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Valérie Lorin
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Oxana Vratskikh
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Thérèse Couderc
- Biology of Infection Unit, INSERM U1117, Department of Cell Biology and Infection, Institut Pasteur, Paris 75015, France
| | - Timothée Bruel
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Paris 75015, France; CNRS URA3015, Paris 75015, France
| | - Olivier Schwartz
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Paris 75015, France; CNRS URA3015, Paris 75015, France
| | | | | | - Jordan D Dimitrov
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France.
| | - Hugo Mouquet
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France.
| |
Collapse
|
33
|
Platt JL, Garcia de Mattos Barbosa M, Cascalho M. The five dimensions of B cell tolerance. Immunol Rev 2019; 292:180-193. [PMID: 31609002 PMCID: PMC10387221 DOI: 10.1111/imr.12813] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
B cell tolerance has been generally understood to be an acquired property of the immune system that governs antibody specificity in ways that avoid auto-toxicity. As useful as this understanding has proved, it fails to fully explain the existence of auto-reactive specificities in healthy individuals and contribution these may have to health. Mechanisms underlying B cell tolerance are considered to select a clonal repertoire that generates a collection of antibodies that do not bind self, ie tolerance operates more or less in three dimensions that largely spare autologous cells and antigens. Yet, most B lymphocytes in humans and probably in other vertebrates are auto-reactive and absence of these auto-reactive B cells is associated with disease. We suggest that auto-reactivity can be embodied by extending the concept of tolerance by two further dimensions, one of time and circumstance and one that allows healthy cells to actively resist injury. In this novel concept, macromolecular recognition by the B cell receptor leading to deletion, anergy, receptor editing or B cell activation is extended by taking account of the time of development of normal immune responses (4th dimension) and the accommodation (or tolerance) of normal cells to bound antibody, activation of complement, and interaction with inflammatory cells (fifth dimension). We discuss how these dimensions contribute to understanding B cell biology in health or disease.
Collapse
Affiliation(s)
- Jeffrey L. Platt
- Department of Surgery University of Michigan Ann Arbor MI USA
- Department of Microbiology and Immunology and Department of Surgery University of Michigan Ann Arbor MI USA
- Lead Contacts Ann Arbor MI USA
| | | | - Marilia Cascalho
- Department of Surgery University of Michigan Ann Arbor MI USA
- Department of Microbiology and Immunology and Department of Surgery University of Michigan Ann Arbor MI USA
- Lead Contacts Ann Arbor MI USA
| |
Collapse
|
34
|
Vanhee S, Åkerstrand H, Kristiansen TA, Datta S, Montano G, Vergani S, Lang S, Ungerbäck J, Doyle A, Olsson K, Beneventi G, Jensen CT, Bellodi C, Soneji S, Sigvardsson M, Gyllenbäck EJ, Yuan J. Lin28b controls a neonatal to adult switch in B cell positive selection. Sci Immunol 2019; 4:4/39/eaax4453. [DOI: 10.1126/sciimmunol.aax4453] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/27/2019] [Indexed: 12/21/2022]
Abstract
The ability of B-1 cells to become positively selected into the mature B cell pool, despite being weakly self-reactive, has puzzled the field since its initial discovery. Here, we explore changes in B cell positive selection as a function of developmental time by exploiting a link between CD5 surface levels and the natural occurrence of self-reactive B cell receptors (BCRs) in BCR wild-type mice. We show that the heterochronic RNA binding protein Lin28b potentiates a neonatal mode of B cell selection characterized by enhanced overall positive selection in general and the developmental progression of CD5+immature B cells in particular. Lin28b achieves this by amplifying the CD19/PI3K/c-Myc positive feedback loop, and ectopic Lin28b expression restores both positive selection and mature B cell numbers in CD19−/−adult mice. Thus, the temporally restricted expression ofLin28brelaxes the rules for B cell selection during ontogeny by modulating tonic signaling. We propose that this neonatal mode of B cell selection represents a cell-intrinsic cue to accelerate the de novo establishment of the adaptive immune system and incorporate a layer of natural antibody-mediated immunity throughout life.
Collapse
|
35
|
Klasse PJ. Non-cognate ligands of Procrustean paratopes as potential vaccine components. EBioMedicine 2019; 47:6-7. [PMID: 31544771 PMCID: PMC6796551 DOI: 10.1016/j.ebiom.2019.07.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 11/05/2022] Open
Affiliation(s)
- P J Klasse
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
36
|
Chiman K, Gholamreza K, Shahram J, Mohammad KB, Reza T, Maryam T, Haghighi SB, Makvandi M. Immuno- and bio-informatic analysis of hexon protein in human adenovirus D8 isolated from patients with keratoconjunctivitis. Future Virol 2019. [DOI: 10.2217/fvl-2018-0165] [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
Aim: In silico analysis of the hexon protein of human adenovirus serotype D-8 isolated from a patients with keratoconjunctivitis in Iran. Materials & methods: The hexon gene of HAdV-D8 was amplified by PCR. HAdV-D8 recovered from EKC outbreak was isolated by growing in A549 cells. Results: The hexon gene isolated from a patient with EKC comprised 2829 nt and 942 aa. The analyses of selected B-cell epitopes prediction (KTFQPEPQIGENNWQD) and T-cell epitopes prediction (TENFDIDLAFFDIPQ), showed high score immunogenicity, which may prove this to be a promising candidate for epitope vaccine development. Conclusion: In silico analysis of selected B-cell epitopes prediction (KTFQPEPQIGENNWQD) and T-cell epitopes prediction (TENFDIDLAFFDIPQ) are immunogenic and provoke B- and T-cell responses.
Collapse
Affiliation(s)
- Karami Chiman
- Infectious & Tropical Disease Research Center, Health Research Institute, & Department of Virology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Jalilian Shahram
- Infectious & Tropical Disease Research Center, Health Research Institute, & Department of Virology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Karimi B Mohammad
- Department of Medical Biotechnology, Faculty of Advance Medical Sciences, Tabriz University of Medical Sciences, East Azerbaijan, Iran
| | - Taherkhani Reza
- Department of Microbiology & Parasitology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Tabasi Maryam
- Infectious & Tropical Disease Research Center, Health Research Institute, & Department of Virology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh B Haghighi
- Department of General Courses, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Manoochehr Makvandi
- Infectious & Tropical Disease Research Center, Health Research Institute, & Department of Virology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
37
|
Dashti A, DeVico AL, Lewis GK, Sajadi MM. Broadly Neutralizing Antibodies against HIV: Back to Blood. Trends Mol Med 2019; 25:228-240. [PMID: 30792120 DOI: 10.1016/j.molmed.2019.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 12/26/2022]
Abstract
After years of continuous exposure to HIV envelope antigens, a minority of HIV-infected individuals develop a cognate polyclonal humoral response comprising very potent and extremely cross-reactive neutralizing antibodies [broadly neutralizing antibodies (bNAbs)]. Isolated bNAbs derived from memory B cell pools have been the focus of intense studies over the past decade. However, it is not yet known how to translate the features of bNAbs into practical HIV prevention methods. In this review, we attempt to seek insights from emerging information about the human broadly neutralizing plasma response as well as its frequency, clonal composition, specificity, potency, and commonality among infected subjects. We also consider how this information points to selecting and prioritizing certain epitope targets and strategies for HIV vaccine design.
Collapse
Affiliation(s)
- Amir Dashti
- Divisions of Vaccine Research and Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Anthony L DeVico
- Divisions of Vaccine Research and Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - George K Lewis
- Divisions of Vaccine Research and Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mohammad M Sajadi
- Divisions of Vaccine Research and Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Medicine, Baltimore VA Medical Center, Baltimore, MD 21201, USA.
| |
Collapse
|
38
|
|
39
|
Sato T, Takano R, Takahara N, Tokunaka K, Saiga K, Tomura A, Sugihara H, Hayashi T, Imamura Y, Morita M. Identification of a common epitope in the sequences of COL4A1 and COL6A1 recognized by monoclonal antibody #141. J Biochem 2019; 165:85-95. [PMID: 30321347 DOI: 10.1093/jb/mvy086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/10/2018] [Indexed: 12/23/2022] Open
Abstract
Identification of a type IV collagen α1 polypeptide in non-triple helical form [NTH α1(IV)], possibly involved in angiogenesis, introduces the further possibility of the existence of non-triple helical forms of other collagen chains. We previously reported that an anti-NTH α1(IV) monoclonal antibody #141 recognizes not only NTH α1(IV) but also a novel non-triple helical collagen polypeptide NTH α1(VI) encoded by COL6A1. In this study, we identified the recognition sequence in order to better understand the properties of antibody #141 and provide clues regarding the biological function of the two non-triple helical molecules. Additionally, we determined the common epitope between COL4A1 and COL6A1 as PXXGXPGLRG, with surface plasmon resonance analyses revealing KD values for the COL4A1 epitope as 5.56±1.81×10-9 M and for the COL6A1 epitope as 7.15±0.44×10-10 M. The specific recognition of NTH α1(IV) and NTH α1(VI) by antibody #141 can be explained by the common epitope sequence. Moreover, epitope localization supports previous finding that NTH α1(IV) and NTH α1(VI) differ in conformation from the α1 chains in triple-helical type IV and type VI collagen. These findings suggest that antibody #141 might be useful for diagnosis of type VI collagen myopathies.
Collapse
Affiliation(s)
- Takamichi Sato
- Biological Research Group, Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd, 3-31-12 Shimo, Kita-ku, Tokyo, Japan
| | - Ryo Takano
- Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2655-1 Nakanomachi, Hachioji, Tokyo, Japan
| | - Naoko Takahara
- Biological Research Group, Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd, 3-31-12 Shimo, Kita-ku, Tokyo, Japan
| | - Kazuhiro Tokunaka
- Biological Research Group, Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd, 3-31-12 Shimo, Kita-ku, Tokyo, Japan
| | - Kan Saiga
- Biological Research Group, Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd, 3-31-12 Shimo, Kita-ku, Tokyo, Japan
| | - Arihiro Tomura
- Biological Research Group, Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd, 3-31-12 Shimo, Kita-ku, Tokyo, Japan
| | - Hidemitsu Sugihara
- Biological Research Group, Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd, 3-31-12 Shimo, Kita-ku, Tokyo, Japan
| | - Toshihiko Hayashi
- Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2655-1 Nakanomachi, Hachioji, Tokyo, Japan.,China-Japan Research Institute of Medical and Pharmaceutical Sciences, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, China
| | - Yasutada Imamura
- Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2655-1 Nakanomachi, Hachioji, Tokyo, Japan
| | - Makoto Morita
- Biological Research Group, Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd, 3-31-12 Shimo, Kita-ku, Tokyo, Japan
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW A successful human immunodeficiency virus-type 1 (HIV-1) vaccine will require immunogens that induce protective immune responses. However, recent studies suggest that the response to HIV-1 and perhaps other viruses may be altered by immune system exposure to intestinal microbiota-antigens. This review will discuss select aspects of these studies. RECENT FINDINGS Naïve CD4 T and B cell repertoires can be imprinted by intestinal microbiota-antigens to respond to virus epitopes prior to virus infection. A multiclade envelope (Env) gp145 DNA prime, recombinant adenovirus type 5 boost vaccine tested in a HIV Vaccine Trials Network (HVTN) phase IIb human vaccine efficacy trial (HVTN 505) induced a dominant gp41-reactive antibody response that was non-neutralizing and cross-reactive with intestinal microbiota. This vaccine regimen also induced a dominant gp41-reactive, intestinal microbiota-cross-reactive gp41 antibody response in neonatal and adult Rhesus macaques. Studies of naïve CD4 T cells have demonstrated cross-reactivity to both HIV-1 and influenza peptides. SUMMARY HIV-1 Env vaccine-induced CD4 T and B cell responses can originate from a pool of intestinal microbiota-cross-reactive immune cells. Moreover, intestinal microbiota-cross-reactive HIV-1 Env antibodies are ineffective in protection against HIV-1 infection. Thus, intestinal microbiota-imprinting of the B cell repertoire may be one of several roadblocks to the induction of protective HIV-1 antibodies.
Collapse
|
41
|
Abstract
Influenza viruses undergo rapid antigenic evolution and reassortment, resulting in annual epidemics and the occasional pandemics. Exposure to influenza virus hemagglutinin (HA) and neuraminidase (NA) antigen, either through vaccination or infection, induces an antibody response able to recognize only the homologous antigenic subtype. However, atypical antibody responses recognizing non-homologous influenza subtypes have been reported during infection and vaccination. Here, we review the incidence of these phenomena in published literature and discuss the potential mechanisms underlying them.
Collapse
Affiliation(s)
- Dalton Hermans
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Richard J Webby
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sook-San Wong
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
42
|
Planchais C, Hocqueloux L, Ibanez C, Gallien S, Copie C, Surenaud M, Kök A, Lorin V, Fusaro M, Delfau-Larue MH, Lefrou L, Prazuck T, Lévy M, Seddiki N, Lelièvre JD, Mouquet H, Lévy Y, Hüe S. Early Antiretroviral Therapy Preserves Functional Follicular Helper T and HIV-Specific B Cells in the Gut Mucosa of HIV-1-Infected Individuals. THE JOURNAL OF IMMUNOLOGY 2018; 200:3519-3529. [PMID: 29632141 DOI: 10.4049/jimmunol.1701615] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/14/2018] [Indexed: 12/25/2022]
Abstract
HIV-1 infection is associated with B cell dysregulation and dysfunction. In HIV-1-infected patients, we previously reported preservation of intestinal lymphoid structures and dendritic cell maturation pathways after early combination antiretroviral therapy (e-ART), started during the acute phase of the infection, compared with late combination antiretroviral therapy started during the chronic phase. In this study, we investigated whether the timing of combination antiretroviral therapy initiation was associated with the development of the HIV-1-specific humoral response in the gut. The results showed that e-ART was associated with higher frequencies of functional resting memory B cells in the gut. These frequencies correlated strongly with those of follicular Th cells in the gut. Importantly, frequencies of HIV-1 Env gp140-reactive B cells were higher in patients given e-ART, in whom gp140-reactive IgG production by mucosal B cells increased after stimulation. Moreover, IL-21 release by PBMCs stimulated with HIV-1 peptide pools was greater with e-ART than with late combination antiretroviral therapy. Thus, early treatment initiation helps to maintain HIV-1-reactive memory B cells in the gut as well as follicular Th cells, whose role is crucial in the development of potent affinity-matured and broadly neutralizing Abs.
Collapse
Affiliation(s)
- Cyril Planchais
- INSERM U955, équipe 16, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France.,Vaccine Research Institute, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France
| | - Laurent Hocqueloux
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Régional d'Orléans-La Source, Orléans F-45000, France
| | - Clara Ibanez
- INSERM U955, équipe 16, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France.,Vaccine Research Institute, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France
| | - Sébastien Gallien
- Vaccine Research Institute, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France.,Assistance Publique-Hôpitaux de Paris, Service d'Immunologie Clinique, Groupe Henri-Mondor Albert-Chenevier, Créteil F-94010, France
| | - Christiane Copie
- Assistance Publique-Hôpitaux de Paris, Département de Pathologie, Groupe Henri-Mondor Albert-Chenevier, Créteil F-94010, France.,INSERM U955, équipe 9, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France
| | - Mathieu Surenaud
- INSERM U955, équipe 16, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France.,Vaccine Research Institute, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France
| | - Ayrin Kök
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Pasteur Institute, Paris 75015, France.,INSERM U1222, Paris 75015, France
| | - Valérie Lorin
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Pasteur Institute, Paris 75015, France.,INSERM U1222, Paris 75015, France
| | - Mathieu Fusaro
- Assistance Publique-Hôpitaux de Paris, Service d'Immunologie Biologique, Groupe Henri-Mondor Albert-Chenevier, Créteil F-94010, France
| | - Marie-Hélène Delfau-Larue
- Assistance Publique-Hôpitaux de Paris, Service d'Immunologie Biologique, Groupe Henri-Mondor Albert-Chenevier, Créteil F-94010, France
| | - Laurent Lefrou
- Service d'Hépato-Gastro-Entérologie, Centre Hospitalier Régional d'Orléans-La Source, Orléans F-45000, France; and
| | - Thierry Prazuck
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Régional d'Orléans-La Source, Orléans F-45000, France
| | - Michael Lévy
- Assistance Publique-Hôpitaux de Paris, Service d'Hépato-Gastro-Entérologie, Groupe Henri-Mondor Albert-Chenevier, Créteil F-94010, France
| | - Nabila Seddiki
- INSERM U955, équipe 16, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France.,Vaccine Research Institute, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France
| | - Jean-Daniel Lelièvre
- INSERM U955, équipe 16, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France.,Vaccine Research Institute, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France.,Assistance Publique-Hôpitaux de Paris, Service d'Immunologie Clinique, Groupe Henri-Mondor Albert-Chenevier, Créteil F-94010, France
| | - Hugo Mouquet
- Vaccine Research Institute, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France.,Laboratory of Humoral Response to Pathogens, Department of Immunology, Pasteur Institute, Paris 75015, France.,INSERM U1222, Paris 75015, France
| | - Yves Lévy
- INSERM U955, équipe 16, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France; .,Vaccine Research Institute, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France.,Assistance Publique-Hôpitaux de Paris, Service d'Immunologie Clinique, Groupe Henri-Mondor Albert-Chenevier, Créteil F-94010, France
| | - Sophie Hüe
- INSERM U955, équipe 16, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France; .,Vaccine Research Institute, Faculté de Médecine, Université Paris Est Créteil, Créteil F-94010, France.,Assistance Publique-Hôpitaux de Paris, Service d'Immunologie Biologique, Groupe Henri-Mondor Albert-Chenevier, Créteil F-94010, France
| |
Collapse
|
43
|
Liberati D, Marzinotto I, Brigatti C, Dugnani E, Pasquale V, Reni M, Balzano G, Falconi M, Piemonti L, Lampasona V. No evidence of pancreatic ductal adenocarcinoma specific autoantibodies to Ezrin in a liquid phase LIPS immunoassay. Cancer Biomark 2018; 22:351-357. [PMID: 29660901 DOI: 10.3233/cbm-181218] [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: 11/15/2022]
Abstract
BACKGROUND Sensitive and specific biomarkers of Pancreatic Ductal Adenocarcinoma (PDAC) are desperately needed to allow early diagnosis and improve patient's survival. Ezrin autoantibodies were recently described as present in 93% of PDAC patients and 40% of healthy subjects who later developed PDAC. However, another prospective study failed to replicate these findings. Both studies were based on the use of a solid phase ELISA immunoassay. OBJECTIVE We aimed at re-evaluating the usefulness of Ezrin autoantibodies as PDAC biomarkers using the Luciferase Immuno Precipitation System (LIPS), an alternative immunoassay format that found successful application for the measurement of autoantibodies against pancreatic autoantigens. METHODS We produced a Nanoluciferase™ tagged Ezrin (NLuc-Ezrin). NLuc-Ezrin was then used as antigen in LIPS to test for Ezrin autoantibodies patients affected by PDAC (n= 40), other pancreatic diseases (OPD, n= 50), and healthy controls (n= 60). RESULTS Overall, binding in liquid phase to Ezrin by serum antibodies was rare and low titer. Furthermore, we did not find statistically significant differences in the prevalence of Ezrin autoantibodies between patients affected by either PDAC or OPD compared to control. CONCLUSIONS Our results do not confirm the usefulness of Ezrin autoAbs as biomarker of PDAC.
Collapse
Affiliation(s)
- Daniela Liberati
- Division of Genetics and Cell Biology, Genomic Unit for the Diagnosis of Human Pathologies, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Division of Genetics and Cell Biology, Genomic Unit for the Diagnosis of Human Pathologies, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Ilaria Marzinotto
- Division of Genetics and Cell Biology, Genomic Unit for the Diagnosis of Human Pathologies, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Division of Genetics and Cell Biology, Genomic Unit for the Diagnosis of Human Pathologies, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Cristina Brigatti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Erica Dugnani
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Valentina Pasquale
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Michele Reni
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Gianpaolo Balzano
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Vita-Salute San Raffaele University, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Vita-Salute San Raffaele University, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Vita-Salute San Raffaele University, Italy
| | - Vito Lampasona
- Division of Genetics and Cell Biology, Genomic Unit for the Diagnosis of Human Pathologies, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| |
Collapse
|
44
|
Borhis G, Trovato M, Chaoul N, Ibrahim HM, Richard Y. B-Cell-Activating Factor and the B-Cell Compartment in HIV/SIV Infection. Front Immunol 2017; 8:1338. [PMID: 29163465 PMCID: PMC5663724 DOI: 10.3389/fimmu.2017.01338] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 10/03/2017] [Indexed: 12/12/2022] Open
Abstract
With the goal to design effective HIV vaccines, intensive studies focused on broadly neutralizing antibodies, which arise in a fraction of HIV-infected people. Apart from identifying new vulnerability sites in the viral envelope proteins, these studies have shown that a fraction of these antibodies are produced by self/poly-reactive B-cells. These findings prompted us to revisit the B-cell differentiation and selection process during HIV/SIV infection and to consider B-cells as active players possibly shaping the helper T-cell program within germinal centers (GCs). In this context, we paid a particular attention to B-cell-activating factor (BAFF), a key cytokine in B-cell development and immune response that is overproduced during HIV/SIV infection. As it does in autoimmune diseases, BAFF excess might contribute to the abnormal rescue of self-reactive B-cells at several checkpoints of the B-cell development and impair memory B-cell generation and functions. In this review, we first point out what is known about the functions of BAFF/a proliferation-inducing ligand and their receptors [B-cell maturation, transmembrane activator and CAML interactor (TACI), and BAFF-R], in physiological and pathophysiological settings, in mice and humans. In particular, we highlight recent results on the previously underappreciated regulatory functions of TACI and on the highly regulated production of soluble TACI and BAFF-R that act as decoy receptors. In light of recent data on BAFF, TACI, and BAFF-R, we then revisit the altered phenotypes and functions of B-cell subsets during the acute and chronic phase of HIV/SIV infection. Given the atypical phenotype and reduced functions of memory B-cells in HIV/SIV infection, we particularly discuss the GC reaction, a key checkpoint where self-reactive B-cells are eliminated and pathogen-specific memory B-cells and plasmablasts/cells are generated in physiological settings. Through its capacity to differentially bind and process BAFF-R and TACI on GC B-cells and possibly on follicular helper T-cells, BAFF appears as a key regulator of the physiological GC reaction. Its local excess during HIV/SIV infection could play a key role in B-cell dysregulations.
Collapse
Affiliation(s)
- Gwenoline Borhis
- INSERM u1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris-Descartes, Paris, France
| | - Maria Trovato
- INSERM u1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris-Descartes, Paris, France
| | - Nada Chaoul
- Commissariat à l’Energie Atomique, Institut des maladies Emergentes et Thérapies innovantes, Service d’Immuno-Virologie, Fontenay-aux Roses, France
| | - Hany M. Ibrahim
- INSERM u1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris-Descartes, Paris, France
| | - Yolande Richard
- INSERM u1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris-Descartes, Paris, France
| |
Collapse
|
45
|
HIV DNA-Adenovirus Multiclade Envelope Vaccine Induces gp41 Antibody Immunodominance in Rhesus Macaques. J Virol 2017; 91:JVI.00923-17. [PMID: 28794027 DOI: 10.1128/jvi.00923-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/31/2017] [Indexed: 11/20/2022] Open
Abstract
Dominant antibody responses in vaccinees who received the HIV-1 multiclade (A, B, and C) envelope (Env) DNA/recombinant adenovirus virus type 5 (rAd5) vaccine studied in HIV-1 Vaccine Trials Network (HVTN) efficacy trial 505 (HVTN 505) targeted Env gp41 and cross-reacted with microbial antigens. In this study, we asked if the DNA/rAd5 vaccine induced a similar antibody response in rhesus macaques (RMs), which are commonly used as an animal model for human HIV-1 infections and for testing candidate HIV-1 vaccines. We also asked if gp41 immunodominance could be avoided by immunization of neonatal RMs during the early stages of microbial colonization. We found that the DNA/rAd5 vaccine elicited a higher frequency of gp41-reactive memory B cells than gp120-memory B cells in adult and neonatal RMs. Analysis of the vaccine-induced Env-reactive B cell repertoire revealed that the majority of HIV-1 Env-reactive antibodies in both adult and neonatal RMs were targeted to gp41. Interestingly, a subset of gp41-reactive antibodies isolated from RMs cross-reacted with host antigens, including autologous intestinal microbiota. Thus, gp41-containing DNA/rAd5 vaccine induced dominant gp41-microbiota cross-reactive antibodies derived from blood memory B cells in RMs as observed in the HVTN 505 vaccine efficacy trial. These data demonstrated that RMs can be used to investigate gp41 immunodominance in candidate HIV-1 vaccines. Moreover, colonization of neonatal RMs occurred within the first week of life, and immunization of neonatal RMs during this time also induced a dominant gp41-reactive antibody response.IMPORTANCE Our results are critical to current work in the HIV-1 vaccine field evaluating the phenomenon of gp41 immunodominance induced by HIV-1 Env gp140 in RMs and humans. Our data demonstrate that RMs are an appropriate animal model to study this phenomenon and to determine the immunogenicity in new HIV-1 Env trimer vaccine designs. The demonstration of gp41 immunodominance in memory B cells of both adult and neonatal RMs indicated that early vaccination could not overcome gp41 dominant responses.
Collapse
|
46
|
HIV transmitted/founder vaccines elicit autologous tier 2 neutralizing antibodies for the CD4 binding site. PLoS One 2017; 12:e0177863. [PMID: 29020058 PMCID: PMC5636061 DOI: 10.1371/journal.pone.0177863] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/04/2017] [Indexed: 01/16/2023] Open
Abstract
Here we report the construction, antigenicity and initial immunogenicity testing of DNA and modified vaccinia Ankara (MVA) vaccines expressing virus-like particles (VLPs) displaying sequential clade C Envelopes (Envs) that co-evolved with the elicitation of broadly neutralizing antibodies (bnAbs) to the CD4 binding site (CD4bs) in HIV-infected individual CH0505. The VLP-displayed Envs showed reactivity for conformational epitopes displayed on the receptor-binding form of Env. Two inoculations of the DNA-T/F vaccine, followed by 3 inoculations of the MVA-T/F vaccine and a final inoculation of the MVA-T/F plus a gp120-T/F protein vaccine elicited nAb to the T/F virus in 2 of 4 rhesus macaques (ID50 of ~175 and ~30). Neutralizing Ab plateaued at 100% neutralization and mapped to the CD4bs like the bnAbs elicited in CH0505. The nAb did not have breadth for other tier 2 viruses. Immunizations with T/F followed by directed-lineage vaccines, both with and without co-delivery of directed-lineage gp120 boosts, failed to elicit tier 2 neutralizing Ab for the CD4bs. Thus, pulsed exposures to DNA and MVA-expressed VLPs plus gp120 protein of a T/F Env can induce autologous tier 2 nAbs to the CD4bs.
Collapse
|
47
|
Bunker JJ, Erickson SA, Flynn TM, Henry C, Koval JC, Meisel M, Jabri B, Antonopoulos DA, Wilson PC, Bendelac A. Natural polyreactive IgA antibodies coat the intestinal microbiota. Science 2017; 358:science.aan6619. [PMID: 28971969 DOI: 10.1126/science.aan6619] [Citation(s) in RCA: 339] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/19/2017] [Indexed: 12/11/2022]
Abstract
Large quantities of immunoglobulin A (IgA) are constitutively secreted by intestinal plasma cells to coat and contain the commensal microbiota, yet the specificity of these antibodies remains elusive. Here we profiled the reactivities of single murine IgA plasma cells by cloning and characterizing large numbers of monoclonal antibodies. IgAs were not specific to individual bacterial taxa but rather polyreactive, with broad reactivity to a diverse, but defined, subset of microbiota. These antibodies arose at low frequencies among naïve B cells and were selected into the IgA repertoire upon recirculation in Peyer's patches. This selection process occurred independent of microbiota or dietary antigens. Furthermore, although some IgAs acquired somatic mutations, these did not substantially influence their reactivity. These findings reveal an endogenous mechanism driving homeostatic production of polyreactive IgAs with innate specificity to microbiota.
Collapse
Affiliation(s)
- Jeffrey J Bunker
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Steven A Erickson
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Theodore M Flynn
- Biosciences Division, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Carole Henry
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jason C Koval
- Biosciences Division, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Marlies Meisel
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Bana Jabri
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Dionysios A Antonopoulos
- Biosciences Division, Argonne National Laboratory, Argonne, IL 60439, USA.,Department of Medicine, University of Chicago, Chicago, IL 60637, USA.,Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Patrick C Wilson
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Albert Bendelac
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA. .,Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
48
|
Abstract
Efforts to develop effective antibody therapeutics are frequently hampered by issues such as aggregation and nonspecificity, often only detected in late stages of the development process. In this study, we used a high throughput cross-reactivity assay to select nonspecific clones from a naïve human repertoire scFv library displayed on the surface of yeast. Most antibody families were de-enriched; however, the rarely expressed VH6 family was highly enriched among nonspecific clones, representing almost 90% of isolated clones. Mutational analysis of this family reveals a dominant role of CDRH2 in driving nonspecific binding. Homology modeling of a panel of VH6 antibodies shows a constrained β-sheet structure in CDRH2 that is not present in other families, potentially contributing to nonspecificity of the family. These findings confirm the common decision to exclude VH6 from synthetic antibody libraries, and support VH6 polyreactivity as a possible important role for the family in early ontogeny and cause for its overabundance in cases of some forms of autoimmunity.
Collapse
Affiliation(s)
- Ryan L Kelly
- a Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , U.S.A
| | - Jessie Zhao
- b Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , U.S.A
| | - Doris Le
- b Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , U.S.A
| | - K Dane Wittrup
- a Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , U.S.A.,b Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , U.S.A
| |
Collapse
|
49
|
Mahmud MN, Oda M, Usui D, Inoshima Y, Ishiguro N, Kamatari YO. A multispecific monoclonal antibody G2 recognizes at least three completely different epitope sequences with high affinity. Protein Sci 2017; 26:2162-2169. [PMID: 28791742 DOI: 10.1002/pro.3263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/18/2017] [Accepted: 07/29/2017] [Indexed: 11/09/2022]
Abstract
A monoclonal antibody (mAb) G2 possesses an unusual characteristic of reacting with at least three proteins (ATP6V1C1, SEPT3, and C6H10orf76) other than its original antigen, chicken prion protein (ChPrP). The epitopes on ChPrP and ATP6V1C1 have been identified previously. In this study, we identified the epitope in the third protein, SEPT3. Interestingly, there was no amino acid sequence similarity among the epitopes on the three proteins. These epitopes had high binding affinities to G2 (KD = ∼10-7 M for monovalent binding and KD = ∼10-9 M for divalent binding), as determined using a SPR biosensor. This is the first report on a three-in-one mAb recognizing completely different epitope sequences with high affinity. Additionally, competitive ELISA indicated that the binding sites on G2, specific for the three different epitopes, overlapped, suggesting that the antigen-binding site may be flexible in the free form and capable of adapting to at least three different conformations to enable interactions with three different antigens.
Collapse
Affiliation(s)
- Md Nuruddin Mahmud
- The United Graduate School of Veterinary Sciences, Gifu University, Gifu, 501-1193, Japan
| | - Masayuki Oda
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Shimogamo, Sakyo-ku, Kyoto, 606-8522, Japan
| | - Daiki Usui
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Shimogamo, Sakyo-ku, Kyoto, 606-8522, Japan
| | - Yasuo Inoshima
- The United Graduate School of Veterinary Sciences, Gifu University, Gifu, 501-1193, Japan.,Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, 501-1193, Japan
| | - Naotaka Ishiguro
- The United Graduate School of Veterinary Sciences, Gifu University, Gifu, 501-1193, Japan.,Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, 501-1193, Japan
| | - Yuji O Kamatari
- Life Science Research Center, Gifu University, Gifu, 501-1193, Japan
| |
Collapse
|
50
|
Nasir IA, Emeribe AU, Shuwa HA, Zakari MM, Peters NO. Type 1 diabetes mellitus and enterovirus linkage: search for associated etiopathology. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2017. [DOI: 10.4103/ejim.ejim_25_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|