Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.378
Filter
Add more filters

Publication year range
1.
Cell ; 183(2): 308-314, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33064987

ABSTRACT

The 2020 Lasker Awards, a celebration of one of the most prestigious international prizes given to individuals for extraordinary contributions to Basic and Clinical Medical Research, Pubic Health, and Special Achievement, was cancelled because of the COVID-19 pandemic. Typically, essays on the awardees and their scientific and medical contributions are solicited and published in Cell in collaboration with the Lasker Committee. This year, the Lasker Committee commissioned an essay to reflect on the historic contributions that scientists and physicians have made to our understanding of immunology and virology, and future directions in medical and basic research that have been highlighted by COVID-19 pandemic.


Subject(s)
Allergy and Immunology , Betacoronavirus/immunology , Coronavirus Infections/immunology , Immunity , Pneumonia, Viral/immunology , Allergy and Immunology/history , Animals , Awards and Prizes , COVID-19 , Cytokines/immunology , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Immune System/cytology , Immunoglobulins/genetics , Immunoglobulins/immunology , Lymphocytes/cytology , Pandemics , SARS-CoV-2 , Vaccination/history
2.
Annu Rev Immunol ; 30: 115-48, 2012.
Article in English | MEDLINE | ID: mdl-22224780

ABSTRACT

Only a handful of the more than 100,000 fungal species on our planet cause disease in humans, yet the number of life-threatening fungal infections in patients has recently skyrocketed as a result of advances in medical care that often suppress immunity intensely. This emerging crisis has created pressing needs to clarify immune defense mechanisms against fungi, with the ultimate goal of therapeutic applications. Herein, we describe recent insights in understanding the mammalian immune defenses deployed against pathogenic fungi. The review focuses on adaptive immune responses to the major medically important fungi and emphasizes how dendritic cells and subsets in various anatomic compartments respond to fungi, recognize their molecular patterns, and signal responses that nurture and shape the differentiation of T cell subsets and B cells. Also emphasized is how the latter deploy effector and regulatory mechanisms that eliminate these nasty invaders while also constraining collateral damage to vital tissue.


Subject(s)
Adaptive Immunity , Fungi/immunology , Mycoses/immunology , Animals , Cell Differentiation/immunology , Chemokines/immunology , Cytokines/immunology , Dendritic Cells/immunology , Humans , Immunity, Innate , Immunoglobulins/immunology , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
Annu Rev Immunol ; 28: 1-19, 2010.
Article in English | MEDLINE | ID: mdl-19968560

ABSTRACT

This article outlines my early start in medicine, a late start in immunology research, and my efforts to integrate the two activities. I first describe some of the background information, excitement, and implications of the recognition of T and B cells as separate but functionally intertwined arms of the adaptive immune system. The article continues with a brief account of my colleagues' and my efforts to use the model of hematopoietic stem cell differentiation along T and B cell lines to gain a better understanding of immunodeficiency diseases and lymphoid malignancies. It concludes with the discovery of a more ancient adaptive immune system in which T-like and B-like cells in jawless vertebrates use variable lymphocyte receptors constructed with leucine-rich-repeat sequences to recognize antigens.


Subject(s)
Adaptive Immunity , B-Lymphocytes/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/cytology , Cell Differentiation , Cell Lineage , Humans , Immunoglobulins/immunology , T-Lymphocytes/cytology
4.
Annu Rev Immunol ; 28: 389-411, 2010.
Article in English | MEDLINE | ID: mdl-20307212

ABSTRACT

B and T lymphocyte associated (BTLA) is an Ig domain superfamily protein with cytoplasmic immunoreceptor tyrosine-based inhibitory motifs. Its ligand, herpesvirus entry mediator (HVEM), is a tumor necrosis factor receptor superfamily member. The unique interaction between BTLA and HVEM allows for a system of bidirectional signaling that must be appropriately regulated to balance the outcome of the immune response. HVEM engagement of BTLA produces inhibitory signals through SH2 domain-containing protein tyrosine phosphatase 1 (Shp-1) and Shp-2 association, whereas BTLA engagement of HVEM produces proinflammatory signals via activation of NF-kappaB. The BTLA-HVEM interaction is intriguing and quite complex given that HVEM has four other ligands that also influence immune responses, the conventional TNF ligand LIGHT and lymphotoxin alpha, as well as herpes simplex virus glycoprotein D and the glycosylphosphatidylinositol-linked Ig domain protein CD160. BTLA-HVEM interactions have been shown to regulate responses in several pathogen and autoimmune settings, but our understanding of this complex system of interactions is certainly incomplete. Recent findings of spontaneous inflammation in BTLA-deficient mice may provide an important clue.


Subject(s)
Glycoproteins/immunology , Receptors, Immunologic/immunology , Receptors, Tumor Necrosis Factor, Member 14/immunology , Animals , Glycoproteins/metabolism , Humans , Immunoglobulins/immunology , Ligands , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Signal Transduction
5.
Nat Immunol ; 17(3): 323-30, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26779600

ABSTRACT

Plasma cell differentiation requires silencing of B cell transcription, while it establishes antibody-secretory function and long-term survival. The transcription factors Blimp-1 and IRF4 are essential for the generation of plasma cells; however, their function in mature plasma cells has remained elusive. We found that while IRF4 was essential for the survival of plasma cells, Blimp-1 was dispensable for this. Blimp-1-deficient plasma cells retained their transcriptional identity but lost the ability to secrete antibody. Blimp-1 regulated many components of the unfolded protein response (UPR), including XBP-1 and ATF6. The overlap in the functions of Blimp-1 and XBP-1 was restricted to that response, with Blimp-1 uniquely regulating activity of the kinase mTOR and the size of plasma cells. Thus, Blimp-1 was required for the unique physiological ability of plasma cells that enables the secretion of protective antibody.


Subject(s)
Cell Differentiation/immunology , Immunoglobulins/immunology , Interferon Regulatory Factors/immunology , Plasma Cells/immunology , Transcription Factors/immunology , Unfolded Protein Response/immunology , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/immunology , Animals , Cell Size , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Immunoglobulins/metabolism , Interferon Regulatory Factors/genetics , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Regulatory Factor X Transcription Factors , Sequence Analysis, DNA , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , Transcription Factors/genetics , Unfolded Protein Response/genetics , X-Box Binding Protein 1
6.
Nat Immunol ; 16(3): 296-305, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25642821

ABSTRACT

Effective vaccines induce high-affinity memory B cells and durable antibody responses through accelerated mechanisms of natural selection. Secondary changes in antibody repertoires after vaccine boosts suggest progressive rediversification of B cell receptors (BCRs), but the underlying mechanisms remain unresolved. Here, the integrated specificity and function of individual memory B cell progeny revealed ongoing evolution of polyclonal antibody specificities through germinal center (GC)-specific transcriptional activity. At the clonal and subclonal levels, single-cell expression of the genes encoding the costimulatory molecule CD83 and the DNA polymerase Polη segregated the secondary GC transcriptional program into four stages that regulated divergent mechanisms of memory BCR evolution. Our studies demonstrate that vaccine boosts reactivate a cyclic program of GC function in class-switched memory B cells to remodel existing antibody specificities and enhance durable immunological protection.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Immunologic Memory/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Antibodies/immunology , Antibody Formation/immunology , Antigens, CD/immunology , DNA-Directed DNA Polymerase/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulins/immunology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Transcription, Genetic/immunology , CD83 Antigen
7.
Nat Immunol ; 14(4): 380-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23455674

ABSTRACT

The transcription factor Bcl-6 orchestrates germinal center (GC) reactions through its actions in B cells and T cells and regulates inflammatory signaling in macrophages. Here we found that genetic replacement with mutated Bcl6 encoding Bcl-6 that cannot bind corepressors to its BTB domain resulted in disruption of the formation of GCs and affinity maturation of immunoglobulins due to a defect in the proliferation and survival of B cells. In contrast, loss of function of the BTB domain had no effect on the differentiation and function of follicular helper T cells or that of other helper T cell subsets. Bcl6-null mice had a lethal inflammatory phenotype, whereas mice with a mutant BTB domain had normal healthy lives with no inflammation. The repression of inflammatory responses by Bcl-6 in macrophages was accordingly independent of the repressor function of the BTB domain. Bcl-6 thus mediates its actions through lineage-specific biochemical functions.


Subject(s)
Cell Lineage/genetics , Cell Lineage/immunology , Inflammation/genetics , Inflammation/immunology , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , Animals , Antibody Affinity/immunology , B-Lymphocytes/immunology , Cell Survival/genetics , Cell Survival/immunology , Chemokines/immunology , Chemokines/metabolism , Female , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Immunoglobulins/immunology , Immunoglobulins/metabolism , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Phenotype , Proto-Oncogene Proteins c-bcl-6/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
8.
Proc Natl Acad Sci U S A ; 119(13): e2117770119, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35312359

ABSTRACT

Spirochetal pathogens, such as the causative agent of Lyme disease, Borrelia burgdorferi sensu lato, encode an abundance of lipoproteins; however, due in part to their evolutionary distance from more well-studied bacteria, such as Proteobacteria and Firmicutes, few spirochetal lipoproteins have assigned functions. Indeed, B. burgdorferi devotes almost 8% of its genome to lipoprotein genes and interacts with its environment primarily through the production of at least 80 surface-exposed lipoproteins throughout its tick vector­vertebrate host lifecycle. Several B. burgdorferi lipoproteins have been shown to serve roles in cellular adherence or immune evasion, but the functions for most B. burgdorferi surface lipoproteins remain unknown. In this study, we developed a B. burgdorferi lipoproteome screening platform utilizing intact spirochetes that enables the identification of previously unrecognized host interactions. As spirochetal survival in the bloodstream is essential for dissemination, we targeted our screen to C1, the first component of the classical (antibody-initiated) complement pathway. We identified two high-affinity C1 interactions by the paralogous lipoproteins, ElpB and ElpQ (also termed ErpB and ErpQ, respectively). Using biochemical, microbiological, and biophysical approaches, we demonstrate that ElpB and ElpQ bind the activated forms of the C1 proteases, C1r and C1s, and represent a distinct mechanistic class of C1 inhibitors that protect the spirochete from antibody-mediated complement killing. In addition to identifying a mode of complement inhibition, our study establishes a lipoproteome screening methodology as a discovery platform for identifying direct host­pathogen interactions that are central to the pathogenesis of spirochetes, such as the Lyme disease agent.


Subject(s)
Bacterial Proteins , Borrelia burgdorferi , Complement C1q , Immune Evasion , Lipoproteins , Lyme Disease , Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Complement C1q/immunology , Humans , Immunoglobulins/immunology , Lipoproteins/immunology , Lyme Disease/immunology , Lyme Disease/microbiology , Proteome/immunology
9.
Hepatology ; 77(3): 965-981, 2023 03 01.
Article in English | MEDLINE | ID: mdl-35938354

ABSTRACT

BACKGROUND AND AIMS: Chronic HBV infection is the leading cause of HCC and a serious health problem in China, East Asia, and North African countries. Effective treatment of HBV-related HCC is currently unavailable. This study evaluated the therapeutic potential of T-cell immunoreceptor with Ig and ITIM domains (TIGIT) blockade in HBV-related HCC. APPROACH AND RESULTS: A mouse model of spontaneous HBV-related HCC was generated by replacing wild-type hepatocytes with HBsAg + hepatocytes (namely HBs-HepR mice). The tumors in HBs-HepR mice were inflammation-associated HCC, similar to HBV-related HCC in patients, which was distinguished from other HCC mouse models, such as diethylnitrosamine-induced HCC, TGF-ß-activated kinase 1 knockout-induced HCC, HCC in a stelic animal model, or NASH-induced HCC. HCC in HBs-HepR mice was characterized by an increased number of CD8 + T cells, whereas the production of IL-2, TNF-α, and interferon-gamma (IFN-γ) by intrahepatic CD8 + T cells was decreased. Increased expression of TIGIT on CD8 + T cells was responsible for functional exhaustion. The therapeutic effect of TIGIT blockade was investigated at the early and middle stages of HCC progression in HBs-HepR mice. TIGIT blockade reinvigorated intrahepatic CD8 + T cells with increased TNF-α and IFN-γ production and an increased number of CD8 + T cells in tumors, thereby slowing the development of HCC in HBs-HepR mice. Blocking PD-L1 did not show direct therapeutic effects or synergize with TIGIT blockade. CONCLUSIONS: Blockade of TIGIT alone enhanced the antitumor activity of CD8 + T cells during the progression of HBV-related HCC in a spontaneous HCC mouse model.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Carcinoma, Hepatocellular/pathology , CD8-Positive T-Lymphocytes , Hepatitis B virus , Liver Neoplasms/pathology , Programmed Cell Death 1 Receptor , Receptors, Antigen, T-Cell/metabolism , Tumor Necrosis Factor-alpha/metabolism , Immunoglobulins/immunology
10.
J Med Virol ; 96(6): e29738, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38884390

ABSTRACT

Immunocompromised individuals are at significantly elevated risk for severe courses of coronavirus disease 2019 (COVID-19). In addition to vaccination, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) neutralizing antibodies (nAbs) have been applied throughout the pandemic, with time of treatment onset and potency against the currently prevailing virus variant identified as relevant factors for medical benefit. Using data from the European Society for Immunodeficiencies (ESID) registry, the present study evaluated COVID-19 cases in three groups of patients with inborn errors of immunity (IEI; 981 agammaglobulinemia patients on immunoglobulin replacement therapy (IGRT); 8960 non-agammaglobulinemia patients on IGRT; 14 428 patients without IGRT), and the neutralizing capacity of 1100 immunoglobulin lots against SARS-CoV-2 ("Wuhan" and Omicron strains), throughout 3 years. From the first (2020/2021) to the second (2021/2022) cold season, i.e., during the virus drift to the more contagious Omicron variants, an increase in case numbers was recorded that was comparable (~2- to 3-fold) for all three study groups. During the same period, immunoglobulin lots showed a profound nAb increase against the archetypal SARS-CoV-2 strain, yet only low levels of Omicron nAbs. Notably, shortly before the third (2022/2023) cold season, Omicron-neutralizing capacity of released immunoglobulin lots had plateaued at high levels. From the second to the third cold season, COVID-19 cases dropped markedly. While a ~6-fold case reduction was recorded for the groups of non-agammaglobulinemia patients on IGRT and IEI patients not receiving IGRT, the decline was ~30-fold for the group of agammaglobulinemia patients on IGRT. These findings suggest a substantial COVID-19-protective effect of IGRT, at least for distinct groups of antibody-deficient patients.


Subject(s)
Agammaglobulinemia , Antibodies, Neutralizing , Antibodies, Viral , COVID-19 , SARS-CoV-2 , Humans , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Agammaglobulinemia/immunology , Agammaglobulinemia/therapy , COVID-19/immunology , COVID-19/therapy , Male , SARS-CoV-2/immunology , Female , Adult , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Middle Aged , Adolescent , Aged , Young Adult , Child , Child, Preschool , Treatment Outcome , Immunoglobulins/therapeutic use , Immunoglobulins/immunology
11.
J Biomed Sci ; 31(1): 58, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824576

ABSTRACT

BACKGROUND: A vaccine against Trypanosoma cruzi, the agent of Chagas disease, would be an excellent additional tool for disease control. A recombinant vaccine based on Tc24 and TSA1 parasite antigens was found to be safe and immunogenic in naïve macaques. METHODS: We used RNA-sequencing and performed a transcriptomic analysis of PBMC responses to vaccination of naïve macaques after each vaccine dose, to shed light on the immunogenicity of this vaccine and guide the optimization of doses and formulation. We identified differentially expressed genes and pathways and characterized immunoglobulin and T cell receptor repertoires. RESULTS: RNA-sequencing analysis indicated a clear transcriptomic response of PBMCs after three vaccine doses, with the up-regulation of several immune cell activation pathways and a broad non-polarized immune profile. Analysis of the IgG repertoire showed that it had a rapid turnover with novel IgGs produced following each vaccine dose, while the TCR repertoire presented several persisting clones that were expanded after each vaccine dose. CONCLUSIONS: These data suggest that three vaccine doses may be needed for optimum immunogenicity and support the further evaluation of the protective efficacy of this vaccine.


Subject(s)
Chagas Disease , Macaca mulatta , Protozoan Vaccines , Receptors, Antigen, T-Cell , Animals , Chagas Disease/immunology , Chagas Disease/prevention & control , Receptors, Antigen, T-Cell/immunology , Protozoan Vaccines/immunology , Trypanosoma cruzi/immunology , Immunoglobulins/immunology
12.
Am J Obstet Gynecol ; 230(5): B2-B5, 2024 05.
Article in English | MEDLINE | ID: mdl-38417536

ABSTRACT

Guidelines for the management of first-trimester spontaneous and induced abortion vary in terms of rhesus factor D (RhD) testing and RhD immune globulin (RhIg) administration. These existing guidelines are based on limited data that do not convincingly demonstrate the safety of withholding RhIg for first-trimester abortions or pregnancy losses. Given the adverse fetal and neonatal outcomes associated with RhD alloimmunization, prevention of maternal sensitization is essential in RhD-negative patients who may experience subsequent pregnancies. In care settings in which RhD testing and RhIg administration are logistically and financially feasible and do not hinder access to abortion care, we recommend offering both RhD testing and RhIg administration for spontaneous and induced abortion at <12 weeks of gestation in unsensitized, RhD-negative individuals. Guidelines for RhD testing and RhIg administration in the first trimester must balance the prevention of alloimmunization with the individual- and population-level harms of restricted access to abortion.


Subject(s)
Abortion, Induced , Abortion, Spontaneous , Maternal-Fetal Exchange , Female , Pregnancy , Abortion, Spontaneous/immunology , Immunoglobulins/immunology , Rh-Hr Blood-Group System/immunology , Societies, Medical , Time Factors , Humans
13.
Fish Shellfish Immunol ; 151: 109751, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38971349

ABSTRACT

Egg yolk antibodies (IgY) can be prepared in large quantities and economically, and have potential value as polyvalent passive vaccines (against multiple bacteria) in aquaculture. This study prepared live and inactivated Vibrio fluvialis IgY and immunized Carassius auratus prior to infection with V. fluvialis and Aeromonas hydrophila. The results showed that the two IgY antibodies hold effective passive protective rates against V. fluvialis and A. hydrophila in C. auratus. Further, the serum of C. auratus recognized the two bacteria in vitro, with a decrease in the bacteria content of the kidney. The phagocytic activity of C. auratus plasma was enhanced, with a decrease in the expression of inflammatory and antioxidant factors. Pathological sections showed that the kidney, spleen, and intestinal tissue structures were intact, and apoptosis and DNA damage decreased in kidney cells. Moreover, the immunoprotection conferred by the live V. fluvialis IgY was higher than that of the inactivated IgY. Addition, live V. fluvialis immunity induced IgY antibodies against outer membrane proteins of V. fluvialis were more than inactivated V. fluvialis immunity. Furthermore, heterologous immune bacteria will not cause infection, so V. fluvialis can be used to immunize chickens to obtain a large amount of IgY antibody. These findings suggest that the passive immunization effect of live bacterial IgY antibody on fish is significantly better than that of inactivated bacterial antibody, and the live V. fluvialis IgY hold potential value as polyvalent passive vaccines in aquaculture.


Subject(s)
Aeromonas hydrophila , Egg Yolk , Fish Diseases , Immunoglobulins , Vibrio Infections , Vibrio , Animals , Immunoglobulins/immunology , Immunoglobulins/blood , Vibrio Infections/veterinary , Vibrio Infections/immunology , Vibrio Infections/prevention & control , Vibrio/immunology , Fish Diseases/immunology , Fish Diseases/prevention & control , Egg Yolk/immunology , Aeromonas hydrophila/immunology , Goldfish/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Gram-Negative Bacterial Infections/prevention & control , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Immunization, Passive/veterinary , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage
14.
Fish Shellfish Immunol ; 150: 109596, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38692380

ABSTRACT

Streptococcosis, the most common bacterial disease of fish in recent years, is highly infectious and lethal, and has become an important factor hindering the healthy and sustainable development of aquaculture. Chicken egg yolk antibody (IgY) has the advantages of high antigen specificity, inexpensive and easy to obtain, simple preparation, no toxic side effects, and in line with animal welfare, which is a green and safe alternative to antibiotics. In this study, the potential of specific IgY in the treatment of gastrointestinal pathogens was explored by observing the effects of specific IgY on intestinal flora, pathological tissue, apoptosis, oxidative stress, and inflammatory response of tilapia. We used the specific IgY prepared in the early stage to feed tilapia for 10 days, and then the tilapia was challenged with Streptococcus agalactiae. The results showed that feeding IgY before challenge had a small effect on the intestinal flora, and after challenge specific IgY decreased the proportion of Streptococcus and increased the diversity of the intestinal flora; in histopathology, specific IgY decreased tissue damage and maintained the integrity of tissue structure. Further study found that specific IgY can reduce intestinal epithelial cell apoptosis and reduce caspase activity; at the same time, the content of MDA was decreased, and the activities of SOD, CAT, GSH-Px and GR were increased. In addition, specific IgY can down-regulate the expression levels of IL-8 and TNF-α genes and up-regulate the expression levels of IL-10 and TGF-ß. The results of this study showed that specific IgY could improve the intestinal flora of tilapia infected with Streptococcus agalactiae, reduce intestinal cell apoptosis, oxidative stress injury and inflammatory response, thereby reducing tissue damage and protecting the health of tilapia. Overall, specific IgY can be further explored as a potential antibiotic alternative for gastrointestinal pathogen infections.


Subject(s)
Animal Feed , Apoptosis , Chickens , Cichlids , Fish Diseases , Gastrointestinal Microbiome , Immunoglobulins , Intestines , Oxidative Stress , Streptococcal Infections , Streptococcus agalactiae , Animals , Streptococcus agalactiae/physiology , Streptococcal Infections/veterinary , Streptococcal Infections/immunology , Oxidative Stress/drug effects , Apoptosis/drug effects , Immunoglobulins/immunology , Cichlids/immunology , Chickens/immunology , Fish Diseases/immunology , Gastrointestinal Microbiome/drug effects , Animal Feed/analysis , Intestines/immunology , Diet/veterinary , Egg Yolk/immunology , Egg Yolk/chemistry
15.
J Immunol ; 208(2): 371-383, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34965965

ABSTRACT

Monoallelic AgR gene expression underlies specific adaptive immune responses. AgR allelic exclusion is achieved by sequential initiation of V(D)J recombination between alleles and resultant protein from one allele signaling to prevent recombination of the other. The ATM kinase, a regulator of the DNA double-strand break (DSB) response, helps enforce allelic exclusion through undetermined mechanisms. ATM promotes repair of RAG1/RAG2 (RAG) endonuclease-induced DSBs and transduces signals from RAG DSBs during Igk gene rearrangement on one allele to transiently inhibit RAG1 protein expression, Igk accessibility, and RAG cleavage of the other allele. Yet, the relative contributions of ATM functions in DSB repair versus signaling to enforce AgR allelic exclusion remain undetermined. In this study, we demonstrate that inactivation in mouse pre-B cells of the NF-κB essential modulator (Nemo) protein, an effector of ATM signaling, diminishes RAG DSB-triggered repression of Rag1/Rag2 transcription and Igk accessibility but does not result in aberrant repair of RAG DSBs like ATM inactivation. We show that Nemo deficiency increases simultaneous biallelic Igk cleavage in pre-B cells and raises the frequency of B cells expressing Igκ proteins from both alleles. In contrast, the incidence of biallelic Igκ expression is not elevated by inactivation of the SpiC transcriptional repressor, which is induced by RAG DSBs in an ATM-dependent manner and suppresses Igk accessibility. Thus, we conclude that Nemo-dependent, ATM-mediated DNA damage signals enforce Igκ allelic exclusion by orchestrating transient repression of RAG expression and feedback inhibition of additional Igk rearrangements in response to RAG cleavage on one Igk allele.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/genetics , Immunoglobulins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Loss of Heterozygosity/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cells, Cultured , Clonal Anergy/genetics , Clonal Anergy/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Immunoglobulins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , V(D)J Recombination/genetics
16.
Nature ; 557(7706): 570-574, 2018 05.
Article in English | MEDLINE | ID: mdl-29769725

ABSTRACT

Arthritogenic alphaviruses comprise a group of enveloped RNA viruses that are transmitted to humans by mosquitoes and cause debilitating acute and chronic musculoskeletal disease 1 . The host factors required for alphavirus entry remain poorly characterized 2 . Here we use a genome-wide CRISPR-Cas9-based screen to identify the cell adhesion molecule Mxra8 as an entry mediator for multiple emerging arthritogenic alphaviruses, including chikungunya, Ross River, Mayaro and O'nyong nyong viruses. Gene editing of mouse Mxra8 or human MXRA8 resulted in reduced levels of viral infection of cells and, reciprocally, ectopic expression of these genes resulted in increased infection. Mxra8 bound directly to chikungunya virus particles and enhanced virus attachment and internalization into cells. Consistent with these findings, Mxra8-Fc fusion protein or anti-Mxra8 monoclonal antibodies blocked chikungunya virus infection in multiple cell types, including primary human synovial fibroblasts, osteoblasts, chondrocytes and skeletal muscle cells. Mutagenesis experiments suggest that Mxra8 binds to a surface-exposed region across the A and B domains of chikungunya virus E2 protein, which are a speculated site of attachment. Finally, administration of the Mxra8-Fc protein or anti-Mxra8 blocking antibodies to mice reduced chikungunya and O'nyong nyong virus infection as well as associated foot swelling. Pharmacological targeting of Mxra8 could form a strategy for mitigating infection and disease by multiple arthritogenic alphaviruses.


Subject(s)
Chikungunya virus/metabolism , Immunoglobulins/metabolism , Membrane Proteins/metabolism , O'nyong-nyong Virus/metabolism , Receptors, Virus/metabolism , 3T3 Cells , Animals , Antibodies, Blocking/immunology , CRISPR-Cas Systems/genetics , Chikungunya virus/pathogenicity , Chondrocytes/metabolism , Fibroblasts/metabolism , Humans , Immunoglobulins/immunology , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , O'nyong-nyong Virus/pathogenicity , Osteoblasts/metabolism , Receptors, Fc/metabolism , Receptors, Virus/deficiency , Receptors, Virus/genetics
17.
Biol Res ; 57(1): 24, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711133

ABSTRACT

Despite the record speed of developing vaccines and therapeutics against the SARS-CoV-2 virus, it is not a given that such success can be secured in future pandemics. In addition, COVID-19 vaccination and application of therapeutics remain low in developing countries. Rapid and low cost mass production of antiviral IgY antibodies could be an attractive alternative or complementary option for vaccine and therapeutic development. In this article, we rapidly produced SARS-CoV-2 antigens, immunized hens and purified IgY antibodies in 2 months after the SARS-CoV-2 gene sequence became public. We further demonstrated that the IgY antibodies competitively block RBD binding to ACE2, neutralize authentic SARS-CoV-2 virus and effectively protect hamsters from SARS-CoV-2 challenge by preventing weight loss and lung pathology, representing the first comprehensive study with IgY antibodies. The process of mass production can be easily implemented in most developing countries and hence could become a new vital option in our toolbox for combating viral pandemics. This study could stimulate further studies, optimization and potential applications of IgY antibodies as therapeutics and prophylactics for human and animals.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 , Chickens , Egg Yolk , Immunoglobulins , SARS-CoV-2 , Animals , SARS-CoV-2/immunology , Antibodies, Neutralizing/immunology , COVID-19/prevention & control , COVID-19/immunology , Chickens/immunology , Cricetinae , Immunoglobulins/immunology , Egg Yolk/immunology , Antibodies, Viral/immunology , Female , Mesocricetus , COVID-19 Vaccines/immunology
18.
Nucleic Acids Res ; 50(D1): D1262-D1272, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34875068

ABSTRACT

IMGT®, the international ImMunoGeneTics information system®, http://www.imgt.org/, is at the forefront of the immunogenetics and immunoinformatics fields with more than 30 years of experience. IMGT® makes available databases and tools to the scientific community pertaining to the adaptive immune response, based on the IMGT-ONTOLOGY. We focus on the recent features of the IMGT® databases, tools, reference directories and web resources, within the three main axes of IMGT® research and development. Axis I consists in understanding the adaptive immune response, by deciphering the identification and characterization of the immunoglobulin (IG) and T cell receptor (TR) genes in jawed vertebrates. It is the starting point of the two other axes, namely the analysis and exploration of the expressed IG and TR repertoires based on comparison with IMGT reference directories in normal and pathological situations (Axis II) and the analysis of amino acid changes and functions of 2D and 3D structures of antibody and TR engineering (Axis III).


Subject(s)
Adaptive Immunity/immunology , Databases, Genetic , Immunogenetics , Vertebrates/genetics , Adaptive Immunity/genetics , Animals , Antibodies/classification , Antibodies/immunology , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Vertebrates/immunology
19.
Adv Exp Med Biol ; 1445: 3-10, 2024.
Article in English | MEDLINE | ID: mdl-38967746

ABSTRACT

The canonical theory of immunology stating that "Immunoglobulin (Ig) is produced by B lymphocytes and exerts antibody activity" has been established since the 1970s. However, the discovery of non B cell-derived Igs (non B-Igs), which can exert multiple biological activities in addition to their antibody activities, necessitates a reevaluation of the classic concept of Ig. This has been documented with a number of characteristics related to their structure, modification, genetic regulation as well as the functions associated with clinical conditions, particularly multiple cancers. The discovery of non B-Ig provides us with a new perspective to better understand not only basic immunology, but also various Ig-related clinical manifestations including autoimmune diseases, chronic inflammation, and anaphylaxis. Notably, non B-Ig can directly promote the occurrence of malignant tumours.


Subject(s)
Immunoglobulins , Humans , Immunoglobulins/immunology , Immunoglobulins/genetics , Animals , B-Lymphocytes/immunology , Neoplasms/immunology , Neoplasms/therapy , Autoimmune Diseases/immunology , Inflammation/immunology
20.
Adv Exp Med Biol ; 1445: 137-149, 2024.
Article in English | MEDLINE | ID: mdl-38967756

ABSTRACT

Intestinal epithelium constitutes a barrier to the unrestricted movement of pathogens, and other detrimental substances from the external world (gut lumen) into the interstitial environment. Intestinal epithelial cells obstruct harmful substances passing through the epithelium as a physical and chemical barrier; Moreover, the epithelial cells can express Toll-like receptors (TLRs) and cytokines to exert innate immune function. In addition, high levels of immunoglobulin A (IgA) and other antibodies exist in the intestinal mucosa, maintaining intestinal immune homeostasis in conjunction with intestinal probiotics. Traditionally, these antibodies have been deemed to be secreted by submucosal plasma cells. Nonetheless, in recent years, it has been demonstrated that intestinal epithelial cells produce a substantial amount of Igs, especially IgA or free Ig light chains, which are involved in intestinal immune homeostasis and the survival of normal epithelial cells. Furthermore, mounting evidence affirms that many human carcinoma cells, including colorectal cancer (CRC), can overexpress Igs, particularly IgG. Cancer-derived Igs exhibit a unique V(D)J rearrangement pattern distinct from B cell-derived Ig; moreover, this cancer cell-derived IgG also has a unique sialic acid modification on the 162 site of CH1 domain (SIA-IgG). The SIA-IgG plays a crucial role in promoting cancer initiation, progression, metastasis, and tumour immune escape. Simultaneously, CRC cells can also express free Ig light chains, which promote colitis, colitis-associated colon carcinogenesis, and CRC progression. Therefore, Igs expressed by CRC cells could be a potential target for diagnosing and preventing the transformation of inflammation into cancer, as well as treating CRC.


Subject(s)
Intestinal Mucosa , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , Immunoglobulins/immunology , Immunoglobulins/metabolism , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL