Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.635
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 36: 359-381, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29400985

ABSTRACT

IgA is the dominant immunoglobulin isotype produced in mammals, largely secreted across the intestinal mucosal surface. Although induction of IgA has been a hallmark feature of microbiota colonization following colonization in germ-free animals, until recently appreciation of the function of IgA in host-microbial mutualism has depended mainly on indirect evidence of alterations in microbiota composition or penetration of microbes in the absence of somatic mutations in IgA (or compensatory IgM). Highly parallel sequencing techniques that enable high-resolution analysis of either microbial consortia or IgA sequence diversity are now giving us new perspectives on selective targeting of microbial taxa and the trajectory of IgA diversification according to induction mechanisms, between different individuals and over time. The prospects are to link the range of diversified IgA clonotypes to specific antigenic functions in modulating the microbiota composition, position and metabolism to ensure host mutualism.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunoglobulin A/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Age Factors , Animals , Disease Susceptibility , Host-Pathogen Interactions/immunology , Humans , Intestinal Mucosa/metabolism , Protein Binding
2.
Cell ; 185(26): 4873-4886.e10, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36513064

ABSTRACT

Respiratory syncytial virus (RSV) infection is a major cause of severe lower respiratory tract infection and death in young infants and the elderly. With no effective prophylactic treatment available, current vaccine candidates aim to elicit neutralizing antibodies. However, binding and neutralization have poorly predicted protection in the past, and accumulating data across epidemiologic cohorts and animal models collectively point to a role for additional antibody Fc-effector functions. To begin to define the humoral correlates of immunity against RSV, here we profiled an adenovirus 26 RSV-preF vaccine-induced humoral immune response in a group of healthy adults that were ultimately challenged with RSV. Protection from infection was linked to opsonophagocytic functions, driven by IgA and differentially glycosylated RSV-specific IgG profiles, marking a functional humoral immune signature of protection against RSV. Furthermore, Fc-modified monoclonal antibodies able to selectively recruit effector functions demonstrated significant antiviral control in a murine model of RSV.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Mice , Animals , Respiratory Syncytial Virus Infections/prevention & control , Antibodies, Neutralizing , Antibodies, Viral , Immunoglobulin G , Immunoglobulin Fc Fragments , Viral Fusion Proteins
3.
Cell ; 181(6): 1276-1290.e13, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32402238

ABSTRACT

At the species level, immunity depends on the selection and transmission of protective components of the immune system. A microbe-induced population of RORγ-expressing regulatory T cells (Tregs) is essential in controlling gut inflammation. We uncovered a non-genetic, non-epigenetic, non-microbial mode of transmission of their homeostatic setpoint. RORγ+ Treg proportions varied between inbred mouse strains, a trait transmitted by the mother during a tight age window after birth but stable for life, resistant to many microbial or cellular perturbations, then further transferred by females for multiple generations. RORγ+ Treg proportions negatively correlated with IgA production and coating of gut commensals, traits also subject to maternal transmission, in an immunoglobulin- and RORγ+ Treg-dependent manner. We propose a model based on a double-negative feedback loop, vertically transmitted via the entero-mammary axis. This immunologic mode of multi-generational transmission may provide adaptability and modulate the genetic tuning of gut immune responses and inflammatory disease susceptibility.


Subject(s)
Digestive System/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Disease Susceptibility/immunology , Female , Gastrointestinal Microbiome/immunology , Homeostasis/immunology , Immunoglobulin A/immunology , Inflammation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred NOD , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
4.
Cell ; 183(1): 169-184.e13, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32931734

ABSTRACT

The coronavirus disease 2019 pandemic has made deployment of an effective vaccine a global health priority. We evaluated the protective activity of a chimpanzee adenovirus-vectored vaccine encoding a prefusion stabilized spike protein (ChAd-SARS-CoV-2-S) in challenge studies with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and mice expressing the human angiotensin-converting enzyme 2 receptor. Intramuscular dosing of ChAd-SARS-CoV-2-S induces robust systemic humoral and cell-mediated immune responses and protects against lung infection, inflammation, and pathology but does not confer sterilizing immunity, as evidenced by detection of viral RNA and induction of anti-nucleoprotein antibodies after SARS-CoV-2 challenge. In contrast, a single intranasal dose of ChAd-SARS-CoV-2-S induces high levels of neutralizing antibodies, promotes systemic and mucosal immunoglobulin A (IgA) and T cell responses, and almost entirely prevents SARS-CoV-2 infection in both the upper and lower respiratory tracts. Intranasal administration of ChAd-SARS-CoV-2-S is a candidate for preventing SARS-CoV-2 infection and transmission and curtailing pandemic spread.


Subject(s)
Coronavirus Infections/immunology , Immunogenicity, Vaccine , Pneumonia, Viral/immunology , Viral Vaccines/immunology , Adenoviridae/genetics , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19 , COVID-19 Vaccines , Chlorocebus aethiops , Coronavirus Infections/pathology , Coronavirus Infections/prevention & control , Female , HEK293 Cells , Humans , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Pandemics , Pneumonia, Viral/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vero Cells , Viral Vaccines/administration & dosage
5.
Cell ; 176(3): 610-624.e18, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30612739

ABSTRACT

Plasma cells (PC) are found in the CNS of multiple sclerosis (MS) patients, yet their source and role in MS remains unclear. We find that some PC in the CNS of mice with experimental autoimmune encephalomyelitis (EAE) originate in the gut and produce immunoglobulin A (IgA). Moreover, we show that IgA+ PC are dramatically reduced in the gut during EAE, and likewise, a reduction in IgA-bound fecal bacteria is seen in MS patients during disease relapse. Removal of plasmablast (PB) plus PC resulted in exacerbated EAE that was normalized by the introduction of gut-derived IgA+ PC. Furthermore, mice with an over-abundance of IgA+ PB and/or PC were specifically resistant to the effector stage of EAE, and expression of interleukin (IL)-10 by PB plus PC was necessary and sufficient to confer resistance. Our data show that IgA+ PB and/or PC mobilized from the gut play an unexpected role in suppressing neuroinflammation.


Subject(s)
Immunoglobulin A/metabolism , Interleukin-10/metabolism , Intestines/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Immunoglobulin A/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Neuroimmunomodulation/immunology , Plasma Cells/metabolism
6.
Cell ; 178(5): 1072-1087.e14, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442401

ABSTRACT

Nutritional status potentially influences immune responses; however, how nutritional signals regulate cellular dynamics and functionality remains obscure. Herein, we report that temporary fasting drastically reduces the number of lymphocytes by ∼50% in Peyer's patches (PPs), the inductive site of the gut immune response. Subsequent refeeding seemingly restored the number of lymphocytes, but whose cellular composition was conspicuously altered. A large portion of germinal center and IgA+ B cells were lost via apoptosis during fasting. Meanwhile, naive B cells migrated from PPs to the bone marrow during fasting and then back to PPs during refeeding when stromal cells sensed nutritional signals and upregulated CXCL13 expression to recruit naive B cells. Furthermore, temporal fasting before oral immunization with ovalbumin abolished the induction of antigen-specific IgA, failed to induce oral tolerance, and eventually exacerbated food antigen-induced diarrhea. Thus, nutritional signals are critical in maintaining gut immune homeostasis.


Subject(s)
B-Lymphocytes/physiology , Immunity, Mucosal , Animals , Antigens/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bone Marrow/immunology , Bone Marrow/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Fasting , Gene Expression Regulation , Glycolysis , Immunoglobulin A/metabolism , Male , Mice , Mice, Inbred BALB C , Nutritional Status , Ovalbumin/immunology , Peyer's Patches/immunology , Peyer's Patches/metabolism , Peyer's Patches/pathology , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism , TOR Serine-Threonine Kinases/metabolism
7.
Immunity ; 57(6): 1428-1441.e8, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38723638

ABSTRACT

Induction of commensal-specific immunity contributes to tissue homeostasis, yet the mechanisms underlying induction of commensal-specific B cells remain poorly understood in part due to a lack of tools to identify these cells. Using phage display, we identified segmented filamentous bacteria (SFB) antigens targeted by serum and intestinal antibodies and generated B cell tetramers to track SFB-specific B cells in gut-associated lymphoid tissues. We revealed a compartmentalized response in SFB-specific B cell activation, with a gradient of immunoglobulin A (IgA), IgG1, and IgG2b isotype production along Peyer's patches contrasted by selective production of IgG2b within mesenteric lymph nodes. V(D)J sequencing and monoclonal antibody generation identified somatic hypermutation driven affinity maturation to SFB antigens under homeostatic conditions. Combining phage display and B cell tetramers will enable investigation of the ontogeny and function of commensal-specific B cell responses in tissue immunity, inflammation, and repair.


Subject(s)
B-Lymphocytes , Animals , B-Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Peyer's Patches/immunology , Lymphocyte Activation/immunology , Antigens, Bacterial/immunology , Somatic Hypermutation, Immunoglobulin , Peptide Library , Lymph Nodes/immunology , Cell Surface Display Techniques , Symbiosis/immunology , Immunoglobulin G/immunology , Immunoglobulin A/immunology
8.
Immunity ; 56(11): 2570-2583.e6, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37909039

ABSTRACT

Dimeric IgA (dIgA) can move through cells via the IgA/IgM polymeric immunoglobulin receptor (PIGR), which is expressed mainly on mucosal epithelia. Here, we studied the ability of dIgA to target commonly mutated cytoplasmic oncodrivers. Mutation-specific dIgA, but not IgG, neutralized KRASG12D within ovarian carcinoma cells and expelled this oncodriver from tumor cells. dIgA binding changed endosomal trafficking of KRASG12D from accumulation in recycling endosomes to aggregation in the early/late endosomes through which dIgA transcytoses. dIgA targeting of KRASG12D abrogated tumor cell proliferation in cell culture assays. In vivo, KRASG12D-specific dIgA1 limited the growth of KRASG12D-mutated ovarian and lung carcinomas in a manner dependent on CD8+ T cells. dIgA specific for IDH1R132H reduced colon cancer growth, demonstrating effective targeting of a cytoplasmic oncodriver not associated with surface receptors. dIgA targeting of KRASG12D restricted tumor growth more effectively than small-molecule KRASG12D inhibitors, supporting the potential of this approach for the treatment of human cancers.


Subject(s)
Carcinoma , Immunoglobulin A , Humans , Immunoglobulin A/metabolism , CD8-Positive T-Lymphocytes/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Cytoplasm/metabolism
9.
Immunity ; 56(10): 2373-2387.e8, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37714151

ABSTRACT

Immunoglobulin A (IgA) maintains commensal communities in the intestine while preventing dysbiosis. IgA generated against intestinal microbes assures the simultaneous binding to multiple, diverse commensal-derived antigens. However, the exact mechanisms by which B cells mount broadly reactive IgA to the gut microbiome remains elusive. Here, we have shown that IgA B cell receptor (BCR) is required for B cell fitness during the germinal center (GC) reaction in Peyer's patches (PPs) and for generation of gut-homing plasma cells (PCs). We demonstrate that IgA BCR drove heightened intracellular signaling in mouse and human B cells, and as a consequence, IgA+ B cells received stronger positive selection cues. Mechanistically, IgA BCR signaling offset Fas-mediated death, possibly rescuing low-affinity B cells to promote a broad humoral response to commensals. Our findings reveal an additional mechanism linking BCR signaling, B cell fate, and antibody production location, which have implications for how intestinal antigen recognition shapes humoral immunity.


Subject(s)
B-Lymphocytes , Peyer's Patches , Mice , Humans , Animals , Antigens/metabolism , Receptors, Antigen, B-Cell/metabolism , Immunoglobulin A , Intestinal Mucosa
10.
Immunity ; 56(1): 43-57.e10, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36630917

ABSTRACT

There is growing recognition that regionalization of bacterial colonization and immunity along the intestinal tract has an important role in health and disease. Yet, the mechanisms underlying intestinal regionalization and its dysregulation in disease are not well understood. This study found that regional epithelial expression of the transcription factor GATA4 controls bacterial colonization and inflammatory tissue immunity in the proximal small intestine by regulating retinol metabolism and luminal IgA. Furthermore, in mice without jejunal GATA4 expression, the commensal segmented filamentous bacteria promoted pathogenic inflammatory immune responses that disrupted barrier function and increased mortality upon Citrobacter rodentium infection. In celiac disease patients, low GATA4 expression was associated with metabolic alterations, mucosal Actinobacillus, and increased IL-17 immunity. Taken together, these results reveal broad impacts of GATA4-regulated intestinal regionalization on bacterial colonization and tissue immunity, highlighting an elaborate interdependence of intestinal metabolism, immunity, and microbiota in homeostasis and disease.


Subject(s)
Enterobacteriaceae Infections , GATA4 Transcription Factor , Gastrointestinal Microbiome , Intestinal Mucosa , Animals , Humans , Mice , Actinobacillus , Gastrointestinal Microbiome/immunology , GATA4 Transcription Factor/metabolism , Immunity, Mucosal , Interleukin-17/immunology , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestine, Small , Symbiosis
11.
Immunity ; 55(10): 1829-1842.e6, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36115337

ABSTRACT

The adult immune system consists of cells that emerged at various times during ontogeny. We aimed to define the relationship between developmental origin and composition of the adult B cell pool during unperturbed hematopoiesis. Lineage tracing stratified murine adult B cells based on the timing of output, revealing that a substantial portion originated within a restricted neonatal window. In addition to B-1a cells, early-life time-stamped B cells included clonally interrelated IgA plasma cells in the gut and bone marrow. These were actively maintained by B cell memory within gut chronic germinal centers and contained commensal microbiota reactivity. Neonatal rotavirus infection recruited recurrent IgA clones that were distinct from those arising by infection with the same antigen in adults. Finally, gut IgA plasma cells arose from the same hematopoietic progenitors as B-1a cells during ontogeny. Thus, a complex layer of neonatally imprinted B cells confer unique antibody responses later in life.


Subject(s)
Immunoglobulin A , Microbiota , Animals , B-Lymphocytes , Germinal Center , Mice , Plasma Cells
12.
Immunity ; 52(4): 635-649.e4, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32240600

ABSTRACT

The intestinal microbiota shapes and directs immune development locally and systemically, but little is known about whether commensal microbes in the stomach can impact their immunological microenvironment. Here, we report that group 2 innate lymphoid cells (ILC2s) were the predominant ILC subset in the stomach and show that their homeostasis and effector functions were regulated by local commensal communities. Microbes elicited interleukin-7 (IL-7) and IL-33 production in the stomach, which in turn triggered the propagation and activation of ILC2. Stomach ILC2s were also rapidly induced following infection with Helicobacter pylori. ILC2-derived IL-5 resulted in the production of IgA, which coated stomach bacteria in both specific pathogen-free (SPF) and H. pylori-infected mice. Our study thus identifies ILC2-dependent IgA response that is regulated by the commensal microbiota, which is implicated in stomach protection by eliminating IgA-coated bacteria including pathogenic H. pylori.


Subject(s)
Gastrointestinal Microbiome/immunology , Helicobacter Infections/immunology , Helicobacter pylori/pathogenicity , Immunoglobulin A/biosynthesis , Interleukin-5/immunology , Stomach/immunology , T-Lymphocyte Subsets/immunology , Animals , Cell Lineage/genetics , Cell Lineage/immunology , Female , Gene Expression Regulation , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/growth & development , Helicobacter pylori/immunology , Immunity, Humoral , Immunity, Innate , Interleukin-33/genetics , Interleukin-33/immunology , Interleukin-5/genetics , Interleukin-7/genetics , Interleukin-7/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , Signal Transduction , Stomach/microbiology , Symbiosis/immunology , T-Lymphocyte Subsets/classification
13.
Immunity ; 52(3): 557-570.e6, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32160523

ABSTRACT

The intestine contains some of the most diverse and complex immune compartments in the body. Here we describe a method for isolating human gut-associated lymphoid tissues (GALTs) that allows unprecedented profiling of the adaptive immune system in submucosal and mucosal isolated lymphoid follicles (SM-ILFs and M-ILFs, respectively) as well as in GALT-free intestinal lamina propria (LP). SM-ILF and M-ILF showed distinct patterns of distribution along the length of the intestine, were linked to the systemic circulation through MAdCAM-1+ high endothelial venules and efferent lymphatics, and had immune profiles consistent with immune-inductive sites. IgA sequencing analysis indicated that human ILFs are sites where intestinal adaptive immune responses are initiated in an anatomically restricted manner. Our findings position ILFs as key inductive hubs for regional immunity in the human intestine, and the methods presented will allow future assessment of these compartments in health and disease.


Subject(s)
Adaptive Immunity/immunology , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Intestines/immunology , Lymphoid Tissue/immunology , Adaptive Immunity/genetics , Animals , Flow Cytometry , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastric Mucosa/ultrastructure , Humans , Immunity, Mucosal/genetics , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Intestines/ultrastructure , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphoid Tissue/metabolism , Lymphoid Tissue/ultrastructure , Microscopy, Confocal , Microscopy, Electron, Scanning , Peyer's Patches/immunology , Peyer's Patches/metabolism , Peyer's Patches/ultrastructure , Sequence Analysis, DNA
14.
EMBO J ; 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39060515

ABSTRACT

Dendritic cell (DC) dysfunction is known to exacerbate intestinal pathologies, but the mechanisms compromising DC-mediated immune regulation in this context remain unclear. Here, we show that intestinal dendritic cells from a mouse model of experimental colitis exhibit significant levels of noncanonical NF-κB signaling, which activates the RelB:p52 heterodimer. Genetic inactivation of this pathway in DCs alleviates intestinal pathologies in mice suffering from colitis. Deficiency of RelB:p52 diminishes transcription of Axin1, a critical component of the ß-catenin destruction complex, reinforcing ß-catenin-dependent expression of Raldh2, which imparts tolerogenic DC attributes by promoting retinoic acid synthesis. DC-specific impairment of noncanonical NF-κB signaling leads to increased colonic numbers of Tregs and IgA+ B cells, which promote luminal IgA production and foster eubiosis. Experimentally introduced ß-catenin haploinsufficiency in DCs with deficient noncanonical NF-κB signaling moderates Raldh2 activity, reinstating colitogenic sensitivity in mice. Finally, inflammatory bowel-disease patients also display a deleterious noncanonical NF-κB signaling signature in intestinal DCs. In sum, we establish how noncanonical NF-κB signaling in dendritic cells can subvert retinoic acid synthesis to fuel intestinal inflammation.

15.
Annu Rev Microbiol ; 77: 645-668, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37713459

ABSTRACT

Secretory antibodies are the only component of our adaptive immune system capable of attacking mucosal pathogens topologically outside of our bodies. All secretory antibody classes are (a) relatively resistant to harsh proteolytic environments and (b) polymeric. Recent elucidation of the structure of secretory IgA (SIgA) has begun to shed light on SIgA functions at the nanoscale. We can now begin to unravel the structure-function relationships of these molecules, for example, by understanding how the bent conformation of SIgA enables robust cross-linking between adjacent growing bacteria. Many mysteries remain, such as the structural basis of protease resistance and the role of noncanonical bacteria-IgA interactions. In this review, we explore the structure-function relationships of IgA from the nano- to the metascale, with a strong focus on how the seemingly banal "license to clump" can have potent effects on bacterial physiology and colonization.


Subject(s)
Immunoglobulin A, Secretory , Biological Transport , Structure-Activity Relationship
16.
Immunity ; 51(3): 508-521.e6, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31471109

ABSTRACT

Recent experimental data and clinical, genetic, and transcriptome evidence from patients converge to suggest a key role of interleukin-1ß (IL-1ß) in the pathogenesis of Kawasaki disease (KD). However, the molecular mechanisms involved in the development of cardiovascular lesions during KD vasculitis are still unknown. Here, we investigated intestinal barrier function in KD vasculitis and observed evidence of intestinal permeability and elevated circulating secretory immunoglobulin A (sIgA) in KD patients, as well as elevated sIgA and IgA deposition in vascular tissues in a mouse model of KD vasculitis. Targeting intestinal permeability corrected gut permeability, prevented IgA deposition and ameliorated cardiovascular pathology in the mouse model. Using genetic and pharmacologic inhibition of IL-1ß signaling, we demonstrate that IL-1ß lies upstream of disrupted intestinal barrier function, subsequent IgA vasculitis development, and cardiac inflammation. Targeting mucosal barrier dysfunction and the IL-1ß pathway may also be applicable to other IgA-related diseases, including IgA vasculitis and IgA nephropathy.


Subject(s)
Cardiovascular Diseases/immunology , Immunoglobulin A/immunology , Inflammation/immunology , Intestines/immunology , Animals , Disease Models, Animal , Humans , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Mucocutaneous Lymph Node Syndrome/immunology , Permeability , Signal Transduction/immunology , Vasculitis/immunology
17.
Immunol Rev ; 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39046160

ABSTRACT

The increasing prevalence of food allergy and related pathologies in recent years has underscored the need to understand the factors affecting adverse reactions to food. Food allergy is caused when food-specific IgE triggers the release of histamine from mast cells. However, other food-specific antibody isotypes exist as well, including IgG and IgA. IgA is the main antibody isotype in the gut and mediates noninflammatory reactions to toxins, commensal bacteria, and food antigens. It has also been thought to induce tolerance to food, thus antagonizing the role of food-specific IgE. However, this has remained unclear as food-specific IgA generation is poorly understood. Particularly, the location of IgA induction, the role of T cell help, and the fates of food-specific B cells remain elusive. In this review, we outline what is known about food-specific IgA induction and highlight areas requiring further study. We also explore how knowledge of food-specific IgA induction can be informed by and subsequently contribute to our overall knowledge of gut immunity.

18.
Immunity ; 49(3): 545-559.e5, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30193848

ABSTRACT

Although the mammalian microbiota is well contained within the intestine, it profoundly shapes development and metabolism of almost every host organ. We questioned the range and depth of microbial metabolite penetration into the host, and how this is modulated by intestinal immunity. Chemically identical microbial and host metabolites were distinguished by stable isotope tracing from 13C-labeled live non-replicating Escherichia coli, differentiating 12C host isotopes with high-resolution mass spectrometry. Hundreds of endogenous microbial compounds penetrated 23 host tissues and fluids after intestinal exposure: subsequent 12C host metabolome signatures included lipidemia, reduced glycolysis, and inflammation. Penetrant bacterial metabolites from the small intestine were rapidly cleared into the urine, whereas induced antibodies curtailed microbial metabolite exposure by accelerating intestinal bacterial transit into the colon where metabolite transport mechanisms are limiting. Pervasive penetration of microbial molecules can cause extensive host tissue responses: these are limited by immune and non-immune intestinal mucosal adaptations to the microbiota.


Subject(s)
Antibodies/metabolism , Gastrointestinal Microbiome/physiology , Glycolysis/immunology , Hyperlipidemias/immunology , Inflammation/immunology , Mammals/immunology , Animals , Antibodies/immunology , Carbon Radioisotopes/analysis , Host-Pathogen Interactions , Immunity , Immunoglobulin Heavy Chains/genetics , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
Semin Immunol ; 67: 101757, 2023 05.
Article in English | MEDLINE | ID: mdl-37003056

ABSTRACT

The dynamic and complex community of microbes that colonizes the intestines is composed of bacteria, fungi, and viruses. At the mucosal surfaces, immunoglobulins play a key role in protection against bacterial and fungal pathogens, and their toxins. Secretory immunoglobulin A (sIgA) is the most abundantly produced antibody at the mucosal surfaces, while Immunoglobulin G (IgG) isotypes play a critical role in systemic protection. IgA and IgG antibodies with reactivity to commensal fungi play an important role in shaping the mycobiota and host antifungal immunity. In this article, we review the latest evidence that establishes a connection between commensal fungi and B cell-mediated antifungal immunity as an additional layer of protection against fungal infections and inflammation.


Subject(s)
Antifungal Agents , Immunoglobulin A, Secretory , Humans , Immunoglobulin G , Bacteria , Immunity, Mucosal , Immunoglobulins
20.
Proc Natl Acad Sci U S A ; 121(3): e2315354120, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38194459

ABSTRACT

The emergence of Omicron lineages and descendent subvariants continues to present a severe threat to the effectiveness of vaccines and therapeutic antibodies. We have previously suggested that an insufficient mucosal immunoglobulin A (IgA) response induced by the mRNA vaccines is associated with a surge in breakthrough infections. Here, we further show that the intramuscular mRNA and/or inactivated vaccines cannot sufficiently boost the mucosal secretory IgA response in uninfected individuals, particularly against the Omicron variant. We thus engineered and characterized recombinant monomeric, dimeric, and secretory IgA1 antibodies derived from four neutralizing IgG monoclonal antibodies (mAbs 01A05, rmAb23, DXP-604, and XG014) targeting the receptor-binding domain of the spike protein. Compared to their parental IgG antibodies, dimeric and secretory IgA1 antibodies showed a higher neutralizing activity against different variants of concern (VOCs), in part due to an increased avidity. Importantly, the dimeric or secretory IgA1 form of the DXP-604 antibody significantly outperformed its parental IgG antibody, and neutralized the Omicron lineages BA.1, BA.2, and BA.4/5 with a 25- to 75-fold increase in potency. In human angiotensin converting enzyme 2 (ACE2) transgenic mice, a single intranasal dose of the dimeric IgA DXP-604 conferred prophylactic and therapeutic protection against Omicron BA.5. Thus, dimeric or secretory IgA delivered by nasal administration may potentially be exploited for the treatment and prevention of Omicron infection, thereby providing an alternative tool for combating immune evasion by the current circulating subvariants and, potentially, future VOCs.


Subject(s)
Antibodies, Monoclonal , Immunoglobulin A, Secretory , Animals , Mice , Humans , Immunoglobulin G , Immunoglobulin A , Administration, Intranasal , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL