Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Science ; 368(6495): 1122-1127, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32381589

ABSTRACT

Immunological memory specific to previously encountered antigens is a cardinal feature of adaptive lymphoid cells. However, it is unknown whether innate myeloid cells retain memory of prior antigenic stimulation and respond to it more vigorously on subsequent encounters. In this work, we show that murine monocytes and macrophages acquire memory specific to major histocompatibility complex I (MHC-I) antigens, and we identify A-type paired immunoglobulin-like receptors (PIR-As) as the MHC-I receptors necessary for the memory response. We demonstrate that deleting PIR-A in the recipient or blocking PIR-A binding to donor MHC-I molecules blocks memory and attenuates kidney and heart allograft rejection. Thus, innate myeloid cells acquire alloantigen-specific memory that can be targeted to improve transplant outcomes.


Subject(s)
Graft Rejection/immunology , Histocompatibility Antigens Class I/immunology , Immunity, Innate , Immunologic Memory , Macrophages/immunology , Monocytes/immunology , Receptors, Immunologic/physiology , Animals , Gene Deletion , Graft Rejection/genetics , Heart Transplantation , Kidney Transplantation , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Receptors, Immunologic/genetics
2.
Sci Immunol ; 4(32)2019 02 01.
Article in English | MEDLINE | ID: mdl-30709843

ABSTRACT

Microbiome sequence analyses have suggested that changes in gut bacterial composition are associated with autoimmune disease in humans and animal models. However, little is known of the mechanisms through which the gut microbiota influences autoimmune responses to distant tissues. Here, we evaluated systemic antibody responses against cultured human gut bacterial strains to determine whether observed patterns of anticommensal antibody (ACAb) responses are associated with type 1 diabetes (T1D) in two cohorts of pediatric study participants. In the first cohort, ACAb responses in sera collected from participants within 6 months of T1D diagnosis were compared with age-matched healthy controls and also with patients with recent onset Crohn's disease. ACAb responses against multiple bacterial species discriminated among these three groups. In the second cohort, we asked whether ACAb responses present before diagnosis were associated with later T1D development and with HLA genotype in participants who were discordant for subsequent progression to diabetes. Serum IgG2 antibodies against Roseburia faecis and against a bacterial consortium were associated with future T1D diagnosis in an HLA DR3/DR4 haplotype-dependent manner. These analyses reveal associations between antibody responses to intestinal microbes and HLA-DR genotype and islet autoantibody specificity and with a future diagnosis of T1D. Further, we present a platform to investigate antibacterial antibodies in biological fluids that is applicable to studies of autoimmune diseases and responses to therapeutic interventions.


Subject(s)
Antibody Formation/immunology , Autoimmunity , Diabetes Mellitus, Type 1/blood , Gastrointestinal Microbiome/immunology , HLA-DR3 Antigen/immunology , HLA-DR4 Antigen/immunology , Islets of Langerhans/immunology , Adolescent , Antibodies, Bacterial/immunology , Autoantibodies/immunology , Child , Clostridiales/immunology , Diabetes Mellitus, Type 1/immunology , Female , Follow-Up Studies , Genotype , Haplotypes , Humans , Male , Prognosis
3.
Sci Immunol ; 2(12)2017 Jun 23.
Article in English | MEDLINE | ID: mdl-28783664

ABSTRACT

Mice devoid of T, B, and natural killer (NK) cells distinguish between self and allogeneic nonself despite the absence of an adaptive immune system. When challenged with an allograft, they mount an innate response characterized by accumulation of mature, monocyte-derived dendritic cells (DCs) that produce interleukin-12 and present antigen to T cells. However, the molecular mechanisms by which the innate immune system detects allogeneic nonself to generate these DCs are not known. To address this question, we studied the innate response of Rag2-/- γc-/- mice, which lack T, B, and NK cells, to grafts from allogeneic donors. By positional cloning, we identified that donor polymorphism in the gene encoding signal regulatory protein α (SIRPα) is a key modulator of the recipient's innate allorecognition response. Donors that differed from the recipient in one or both Sirpa alleles elicited an innate alloresponse. The response was mediated by binding of donor SIRPα to recipient CD47 and was modulated by the strength of the SIRPα-CD47 interaction. Therefore, sensing SIRPα polymorphism by CD47 provides a molecular mechanism by which the innate immune system distinguishes between self and allogeneic nonself independently of T, B, and NK cells.

4.
J Immunol ; 194(12): 5663-73, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25964492

ABSTRACT

Type 1 diabetes in the NOD mouse model has been linked to >30 insulin-dependent diabetes (Idd) susceptibility loci. Idd4 on chromosome 11 consists of two subloci, Idd4.1 and Idd4.2. Using congenic analysis of alleles in NOD and NOD-resistant (NOR) mice, we previously defined Idd4.1 as an interval containing >50 genes that controlled expression of genes in the type 1 IFN pathway. In this study, we report refined mapping of Idd4.1 to a 1.1-Mb chromosomal region and provide genomic sequence analysis and mechanistic evidence supporting its role in innate immune regulation of islet-directed autoimmunity. Genetic variation at Idd4.1 was mediated by radiation-sensitive hematopoietic cells, and type 1 diabetes protection conferred by the NOR allele was abrogated in mice treated with exogenous type 1 IFN-ß. Next generation sequence analysis of the full Idd4.1 genomic interval in NOD and NOR strains supported Nlrp1b as a strong candidate gene for Idd4.1. Nlrp1b belongs to the Nod-like receptor (NLR) gene family and contributes to inflammasome assembly, caspase-1 recruitment, and release of IL-1ß. The Nlrp1b of NOR was expressed as an alternative spliced isoform that skips exon 9, resulting in a premature stop codon predicted to encode a truncated protein. Functional analysis of the truncated NOR Nlrp1b protein demonstrated that it was unable to recruit caspase-1 and process IL-1ß. Our data suggest that Idd4.1-dependent protection from islet autoimmunity is mediated by differences in type 1 IFN- and IL-1ß-dependent immune responses resulting from genetic variation in Nlrp1b.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Inflammasomes/genetics , Quantitative Trait Loci , Alleles , Alternative Splicing , Animals , Apoptosis Regulatory Proteins/chemistry , Base Sequence , Chromosome Mapping , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Female , Genetic Association Studies , Inflammasomes/immunology , Interferon-beta/metabolism , Interferon-beta/pharmacology , Male , Mice , Mice, Inbred NOD , Molecular Sequence Data , Protein Interaction Domains and Motifs , Sequence Alignment
5.
J Immunol ; 193(10): 4833-44, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25305319

ABSTRACT

The signal regulatory protein (SIRP) locus encodes a family of paired receptors that mediate both activating and inhibitory signals and is associated with type 1 diabetes (T1D) risk. The NOD mouse model recapitulates multiple features of human T1D and enables mechanistic analysis of the impact of genetic variations on disease. In this study, we identify Sirpa encoding an inhibitory receptor on myeloid cells as a gene in the insulin-dependent diabetes locus 13.2 (Idd13.2) that drives islet inflammation and T1D. Compared to T1D-resistant strains, the NOD variant of SIRPα displayed greater binding to its ligand CD47, as well as enhanced T cell proliferation and diabetogenic potency. Myeloid cell-restricted expression of a Sirpa transgene accelerated disease in a dose-dependent manner and displayed genetic and functional interaction with the Idd5 locus to potentiate insulitis progression. Our study demonstrates that variations in both SIRPα sequence and expression level modulate T1D immunopathogenesis. Thus, we identify Sirpa as a T1D risk gene and provide insight into the complex mechanisms by which disease-associated variants act in concert to drive defined stages in disease progression.


Subject(s)
CD47 Antigen/genetics , Diabetes Mellitus, Type 1/immunology , Immunity, Innate , Polymorphism, Genetic/immunology , Receptors, Immunologic/genetics , Animals , Autoimmunity , CD47 Antigen/immunology , Cell Proliferation , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Gene Expression Regulation , Genetic Loci , Humans , Ligands , Mice , Mice, Inbred NOD , Myeloid Cells/immunology , Myeloid Cells/pathology , Protein Binding , Receptors, Immunologic/immunology , Risk Factors , Signal Transduction , Transgenes
6.
J Exp Med ; 210(12): 2539-52, 2013 Nov 18.
Article in English | MEDLINE | ID: mdl-24145514

ABSTRACT

Prnp(-/-) mice lack the prion protein PrP(C) and are resistant to prion infections, but variable phenotypes have been reported in Prnp(-/-) mice and the physiological function of PrP(C) remains poorly understood. Here we examined a cell-autonomous phenotype, inhibition of macrophage phagocytosis of apoptotic cells, previously reported in Prnp(-/-) mice. Using formal genetic, genomic, and immunological analyses, we found that the regulation of phagocytosis previously ascribed to PrP(C) is instead controlled by a linked locus encoding the signal regulatory protein α (Sirpa). These findings indicate that control of phagocytosis was previously misattributed to the prion protein and illustrate the requirement for stringent approaches to eliminate confounding effects of flanking genes in studies modeling human disease in gene-targeted mice. The plethora of seemingly unrelated functions attributed to PrP(C) suggests that additional phenotypes reported in Prnp(-/-) mice may actually relate to Sirpa or other genetic confounders.


Subject(s)
Phagocytosis/genetics , Phagocytosis/physiology , Prions/genetics , Prions/physiology , Receptors, Immunologic/genetics , Receptors, Immunologic/physiology , Amino Acid Sequence , Animals , Apoptosis , Female , Humans , Macrophages, Peritoneal/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Knockout , Molecular Sequence Data , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Prion Proteins , Sequence Homology, Amino Acid
7.
Science ; 339(6123): 1084-8, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23328391

ABSTRACT

Microbial exposures and sex hormones exert potent effects on autoimmune diseases, many of which are more prevalent in women. We demonstrate that early-life microbial exposures determine sex hormone levels and modify progression to autoimmunity in the nonobese diabetic (NOD) mouse model of type 1 diabetes (T1D). Colonization by commensal microbes elevated serum testosterone and protected NOD males from T1D. Transfer of gut microbiota from adult males to immature females altered the recipient's microbiota, resulting in elevated testosterone and metabolomic changes, reduced islet inflammation and autoantibody production, and robust T1D protection. These effects were dependent on androgen receptor activity. Thus, the commensal microbial community alters sex hormone levels and regulates autoimmune disease fate in individuals with high genetic risk.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1/microbiology , Gonadal Steroid Hormones/immunology , Intestines/microbiology , Metagenome , Sex Characteristics , Animals , Cecum/microbiology , Female , Male , Mice , Mice, Inbred NOD
8.
Nat Immunol ; 8(12): 1313-23, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17982459

ABSTRACT

Graft failure in the transplantation of hematopoietic stem cells occurs despite donor-host genetic identity of human leukocyte antigens, suggesting that additional factors modulate engraftment. With the nobese diabetic (NOD)-severe combined immunodeficiency (SCID) xenotransplantation model, we found that the NOD background allowed better hematopoietic engraftment than did other strains with equivalent immunodeficiency-related mutations. We used positional genetics to characterize the molecular basis for this strain specificity and found that the NOD Sirpa allele conferred support for human hematopoiesis. NOD SIRP-alpha showed enhanced binding to the human CD47 ligand, and its expression on mouse macrophages was required for support of human hematopoiesis. Thus, we have identified Sirpa polymorphism as a potent genetic determinant of the engraftment of human hematopoietic stem cells.


Subject(s)
Antigens, Differentiation/physiology , Hematopoietic Stem Cell Transplantation , Polymorphism, Genetic , Receptors, Immunologic/physiology , Animals , Antigens, Differentiation/genetics , Hematopoietic Stem Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Immunologic/genetics
9.
Diabetes ; 55(12): 3611-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17130511

ABSTRACT

The nonobese diabetic (NOD) mouse recapitulates many aspects of the pathogenesis of type 1 diabetes in humans, including inheritance as a complex trait. More than 20 Idd loci have been linked to type 1 diabetes susceptibility in NOD mice. Previously, we used linkage analysis of NOD crossed to the nonobese diabetes-resistant (NOR) strain and NOD congenic strains to map susceptibility to both spontaneous and cyclophosphamide-accelerated type 1 diabetes to the Idd4 locus on chromosome 11 that displayed a sex-specific effect on diabetes susceptibility. Here, we elucidate the complex genetic architecture of Idd4 by analysis of congenic strains on the NOD and NOR backgrounds. We previously refined Idd4.1 to 1.4 Mb and demonstrated an impact of this interval on type 1 interferon pathways in antigen-presenting cells. Here, we identify a second subregion, the 0.92 Mb Idd4.2 locus located telomeric to Idd4.1. Strikingly, Idd4.2 displayed a sex-specific, epistatic interaction with Idd4.1 in NOR.NOD congenic females that was not observed in syngenic males. Idd4.2 contains 29 genes, and promising candidates for the Idd4.2 effect on type 1 diabetes are described. These data demonstrate sex-dependent interaction effects on type 1 diabetes susceptibility and provide a framework for functional analysis of Idd4.2 candidate genes.


Subject(s)
Chromosome Mapping , Diabetes Mellitus, Type 1/genetics , Mice, Inbred NOD/genetics , Animals , Base Sequence , DNA/genetics , DNA/isolation & purification , DNA Primers , Female , Genetic Predisposition to Disease , Male , Mice , Nitric Oxide Synthase Type II/genetics , Sex Characteristics , Species Specificity
10.
J Immunol ; 176(5): 2976-90, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16493056

ABSTRACT

High-resolution mapping and identification of the genes responsible for type 1 diabetes (T1D) has proved difficult because of the multigenic etiology and low penetrance of the disease phenotype in linkage studies. Mouse congenic strains have been useful in refining Idd susceptibility loci in the NOD mouse model and providing a framework for identification of genes underlying complex autoimmune syndromes. Previously, we used NOD and a nonobese diabetes-resistant strain to map the susceptibility to T1D to the Idd4 locus on chromosome 11. Here, we report high-resolution mapping of this locus to 1.4 megabases. The NOD Idd4 locus was fully sequenced, permitting a detailed comparison with C57BL/6 and DBA/2J strains, the progenitors of T1D resistance alleles found in the nonobese diabetes-resistant strain. Gene expression arrays and quantitative real-time PCR were used to prioritize Idd4 candidate genes by comparing macrophages/dendritic cells from congenic strains where allelic variation was confined to the Idd4 interval. The differentially expressed genes either were mapped to Idd4 or were components of the IFN response pathway regulated in trans by Idd4. Reflecting central roles of Idd4 genes in Ag presentation, arachidonic acid metabolism and inflammation, phagocytosis, and lymphocyte trafficking, our combined analyses identified Alox15, Alox12e, Psmb6, Pld2, and Cxcl16 as excellent candidate genes for the effects of the Idd4 locus.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Genetic Predisposition to Disease , Interferons/physiology , Animals , Chromosome Mapping , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/metabolism , Female , Gene Expression Profiling , Genetic Markers , Genomics , Macrophages/metabolism , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred NOD , Signal Transduction/genetics , Signal Transduction/immunology
11.
J Immunol ; 174(11): 7129-40, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15905556

ABSTRACT

Many human autoimmune diseases are more frequent in females than males, and their clinical severity is affected by sex hormone levels. A strong female bias is also observed in the NOD mouse model of type I diabetes (T1D). In both NOD mice and humans, T1D displays complex polygenic inheritance and T cell-mediated autoimmune pathogenesis. The identities of many of the insulin-dependent diabetes (Idd) loci, their influence on specific stages of autoimmune pathogenesis, and sex-specific effects of Idd loci in the NOD model are not well understood. To address these questions, we analyzed cyclophosphamide-accelerated T1D (CY-T1D) that causes disease with high and similar frequencies in male and female NOD mice, but not in diabetes-resistant animals, including the nonobese diabetes-resistant (NOR) strain. In this study we show by genetic linkage analysis of (NOD x NOR) x NOD backcross mice that progression to severe islet inflammation after CY treatment was controlled by the Idd4 and Idd9 loci. Congenic strains on both the NOD and NOR backgrounds confirmed the roles of Idd4 and Idd9 in CY-T1D susceptibility and revealed the contribution of a third locus, Idd5. Importantly, we show that the three loci acted at distinct stages of islet inflammation and disease progression. Among these three loci, Idd4 alleles alone displayed striking sex-specific behavior in CY-accelerated disease. Additional studies will be required to address the question of whether a sex-specific effect of Idd4, observed in this study, is also present in the spontaneous model of the disease with striking female bias.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/physiopathology , Genetic Predisposition to Disease/genetics , Sex Characteristics , Alleles , Animals , Biotransformation/immunology , Cyclophosphamide/metabolism , Cyclophosphamide/toxicity , Diabetes Mellitus, Type 1/immunology , Female , Genetic Linkage/immunology , Genetic Markers/immunology , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Severity of Illness Index , Species Specificity
12.
Cancer Res ; 62(20): 5828-34, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12384545

ABSTRACT

Transformed lymphocytes exhibit aberrant growth potential resulting from enhanced proliferation and resistance to apoptotic stimuli. These mechanisms also influence the development of autoimmune disease, where dysregulated lymphocyte homeostasis has been implicated in expansion of autoreactive T cells. In the nonobese diabetic (NOD) mouse, a murine model of autoimmune type 1 diabetes and Sjögren's syndrome, T cells are apoptosis resistant compared with other mouse strains, a feature thought to potentiate their autoimmune function. NOD mice congenic for the severe combined immunodeficiency scid mutation (NOD.scid) have an incidence of pro-T-cell lymphoma far in excess of scid mutants on other genetic backgrounds. This mutation arrests lymphocyte development secondary to a generalized defect in double-strand DNA break repair that compromises V(D)J recombination. To distinguish between the contributions of immunodeficiency and defective double-strand DNA break repair to lymphoma susceptibility on the NOD background, we examined the incidence, phenotype, and molecular mechanisms of lymphoma development in two immunodeficient NOD strains with normal DNA repair function. We report that NOD mice deficient in mature B cells (NOD. micro MT) or mature T and B cells (NOD.RAG-2(-/-)) display a high incidence of lymphoma of both T- and B-cell origin compared with these mutations on other genetic backgrounds. Strikingly, the lymphoma incidence in both strains was greater in females, mirroring the greater incidence of autoimmune type 1 diabetes in NOD females than in males. The high incidence of autoimmune diabetes and lymphoma in immunodeficiency NOD mice suggests the presence of genetic modifiers that affect lymphocyte homeostasis.


Subject(s)
Lymphoma/immunology , Severe Combined Immunodeficiency/immunology , Animals , B-Lymphocytes/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/immunology , Female , Genetic Predisposition to Disease , Germ-Line Mutation , Lymphoma/genetics , Male , Mice , Mice, Inbred NOD , Severe Combined Immunodeficiency/genetics , Sex Factors , Splenic Neoplasms/genetics , Splenic Neoplasms/immunology , T-Lymphocytes/immunology , Thymus Neoplasms/genetics , Thymus Neoplasms/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...