Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
Add more filters

Publication year range
1.
J Autoimmun ; 108: 102402, 2020 03.
Article in English | MEDLINE | ID: mdl-31980336

ABSTRACT

We have previously identified a signature HLA-DR3 pocket variant, designated HLA-DRß1-Arg74 that confers a high risk for Graves' Disease (GD). In view of the key role of HLA-DRß1-Arg74 in triggering GD we hypothesized that thyroid-stimulating hormone receptor (TSHR) peptides that bind to the HLA-DRß1-Arg74 pocket with high affinity represent key pathogenic TSHR peptides triggering GD, and that blocking their presentation to CD4+ T-cells can be used as a novel therapeutic approach in GD. There were several previous attempts to identify the major pathogenic TSHR peptide utilizing different methodologies, however the results were inconsistent and inconclusive. Therefore, the aim of our study was to use TSHR peptide binding affinity to HLA-DRß1-Arg74 as a method to identify the key pathogenic TSHR peptides that trigger GD. Using virtual screening and ELISA and cellular binding assays we identified 2 TSHR peptides that bound with high affinity to HLA-DRß1-Arg74 - TSHR.132 and TSHR.197. Peptide immunization studies in humanized DR3 mice showed that only TSHR.132, but not TSHR.197, induced autoreactive T-cell proliferation and cytokine responses. Next, we induced experimental autoimmune Graves' disease (EAGD) in a novel BALB/c-DR3 humanized mouse model we created and confirmed TSHR.132 as a major DRß1-Arg74 binding peptide triggering GD in our mouse model. Furthermore, we demonstrated that Cepharanthine, a compound we have previously identified as DRß1-Arg74 blocker, could block the presentation and T-cell responses to TSHR.132 in the EAGD model.


Subject(s)
Antigen Presentation/drug effects , Antigen Presentation/immunology , Benzylisoquinolines/pharmacology , HLA-DR3 Antigen/immunology , Peptides/antagonists & inhibitors , Peptides/immunology , Receptors, Thyrotropin/immunology , Amino Acid Sequence , Animals , Benzylisoquinolines/chemistry , Epitope Mapping/methods , Epitopes, T-Lymphocyte/immunology , Flow Cytometry , Graves Disease/diagnosis , Graves Disease/drug therapy , Graves Disease/immunology , HLA-DR3 Antigen/genetics , Humans , Immunohistochemistry , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Mice, Transgenic , Models, Molecular , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/immunology , Peptides/chemistry , Protein Binding , Receptors, Thyrotropin/chemistry , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
2.
J Immunol ; 198(11): 4413-4424, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28468970

ABSTRACT

Mature peripheral double negative T (DNT) cells expressing αß TCR but lacking CD4/CD8 coreceptors play protective as well as pathogenic roles. To better understand their development and functioning in vivo, we concomitantly inactivated CD4 and CD8 genes in mice with intact MHC class I and class II molecules with the hypothesis that this would enable the development of DNT cells. We also envisaged that these DNT cells could be activated by bacterial superantigens in vivo as activation of T cells by superantigens does not require CD4 and CD8 coreceptors. Because HLA class II molecules present superantigens more efficiently than murine MHC class II molecules, CD4 CD8 double knockout (DKO) mice transgenically expressing HLA-DR3 or HLA-DQ8 molecules were generated. Although thymic cellularity was comparable between wild type (WT) and DKO mice, CD3+ αß TCR+ thymocytes were significantly reduced in DKO mice, implying defects in thymic-positive selection. Splenic CD3+ αß TCR+ cells and Foxp3+ T regulatory cells were present in DKO mice but significantly reduced. However, the in vivo inflammatory responses and immunopathology elicited by acute challenge with the staphylococcal superantigen enterotoxin B were comparable between WT and DKO mice. Choric exposure to staphylococcal enterotoxin B precipitated a lupus-like inflammatory disease with characteristic lympho-monocytic infiltration in lungs, livers, and kidneys, along with production of anti-nuclear Abs in DKO mice as in WT mice. Overall, our results suggest that DNT cells can develop efficiently in vivo and chronic exposure to bacterial superantigens may precipitate a lupus-like autoimmune disease through activation of DNT cells.


Subject(s)
CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Enterotoxins/immunology , Superantigens/immunology , T-Lymphocyte Subsets/immunology , Animals , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/immunology , Histocompatibility Antigens Class II/immunology , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Spleen/cytology , Spleen/immunology , Thymus Gland/cytology , Thymus Gland/immunology
3.
J Immunol ; 196(8): 3253-63, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26944932

ABSTRACT

Identifying T cell epitopes of islet autoantigens is important for understanding type 1 diabetes (T1D) immunopathogenesis and to design immune monitoring and intervention strategies in relationship to disease progression. Naturally processed T cell epitopes have been discovered by elution from HLA-DR4 of pulsed B lymphocytes. The designated professional APC directing immune responses is the dendritic cell (DC). To identify naturally processed epitopes, monocyte-derived DC were pulsed with preproinsulin (PPI), glutamic acid decarboxylase (65-kDa isoform; GAD65), and insulinoma-associated Ag-2 (IA-2), and peptides were eluted of HLA-DR3 and -DR4, which are associated with highest risk for T1D development. Proteome analysis confirmed uptake and processing of islet Ags by DC. PPI peptides generated by DC differed from those processed by B lymphocytes; PPI signal-sequence peptides were eluted from HLA-DR4 and -DR3/4 that proved completely identical to a primary target epitope of diabetogenic HLA-A2-restricted CD8 T cells. HLA-DR4 binding was confirmed. GAD65 peptides, eluted from HLA-DR3 and -DR4, encompassed two core regions overlapping the two most immunodominant and frequently studied CD4 T cell targets. GAD65 peptides bound to HLA-DR3. Strikingly, the IA-2 ligandome of HLA-DR was exclusively generated from the extracellular part of IA-2, whereas most previous immune studies have focused on intracellular IA-2 epitopes. The newly identified IA-2 peptides bound to HLA-DR3 and -DR4. Differential T cell responses were detected against the newly identified IA-2 epitopes in blood from T1D patients. The core regions to which DC may draw attention from autoreactive T cells are largely distinct and more restricted than are those of B cells. GAD65 peptides presented by DC focus on highly immunogenic T cell targets, whereas HLA-DR-binding peptides derived from IA-2 are distinct from the target regions of IA-2 autoantibodies.


Subject(s)
Autoimmunity/immunology , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , HLA-DR3 Antigen/immunology , HLA-DR4 Antigen/immunology , Islets of Langerhans/immunology , Autoantigens/immunology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Epitopes, T-Lymphocyte/immunology , Glutamate Decarboxylase/metabolism , Humans , Insulin/metabolism , Lymphocyte Activation/immunology , Protein Binding/immunology , Protein Precursors/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 8/metabolism
4.
J Immunol ; 195(10): 4660-7, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26475924

ABSTRACT

MHC, especially HLA-DR3 and HLA-DR2, is one of the most important genetic susceptibility regions for systemic lupus erythematosus. Human studies to understand the role of specific HLA alleles in disease pathogenesis have been hampered by the presence of strong linkage disequilibrium in this region. To overcome this, we produced transgenic mice expressing HLA-DR3 (DRß1*0301) and devoid of endogenous class II (both I-A and I-E genes, AE(0)) on a lupus-prone NZM2328 background (NZM2328.DR3(+)AE(0)). Both NZM2328 and NZM2328.DR3(+)AE(0) mice developed anti-dsDNA and glomerulonephritis, but anti-dsDNA titers were higher in the latter. Although kidney histological scores were similar in NZM2328 and NZM2328.DR3(+)AE(0) mice (7.2 ± 4.3 and 8.6 ± 5.7, respectively, p = 0.48), the onset of severe proteinuria occurred earlier in NZM2328.DR3(+)AE(0) mice compared with NZM2328 mice (median, 5 and 9 mo respectively, p < 0.001). Periarterial lymphoid aggregates, classic wire loop lesions, and occasional crescents were seen only in kidneys from NZM2328.DR3(+)AE(0) mice. Interestingly, NZM2328.DR3(+)AE(0) mice, but not NZM2328 mice, spontaneously developed anti-Smith (Sm) Abs. The anti-Sm Abs were seen in NZM2328.DR3(+)AE(0) mice that were completely devoid of endogenous class II (AE(-/) (-)) but not in mice homozygous (AE(+/+)) or heterozygous (AE(+/-)) for endogenous MHC class II. It appears that only HLA-DR3 molecules can preferentially select SmD-reactive CD4(+) T cells for generation of the spontaneous anti-Sm immune response. Thus, our mouse model unravels a critical role for HLA-DR3 in generating an autoimmune response to SmD and lupus nephritis in the NZM2328 background.


Subject(s)
Antibodies, Antinuclear/immunology , Glomerulonephritis/immunology , HLA-DR3 Antigen/immunology , Lupus Nephritis/immunology , snRNP Core Proteins/immunology , Animals , Antibodies, Antinuclear/genetics , CD4-Positive T-Lymphocytes/immunology , DNA/immunology , Disease Models, Animal , Genetic Predisposition to Disease , Glomerulonephritis/genetics , HLA-DR2 Antigen/immunology , HLA-DR3 Antigen/genetics , HLA-DRB1 Chains/genetics , HLA-DRB1 Chains/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/genetics , Mice , Mice, Knockout
5.
Clin Exp Immunol ; 185(2): 133-40, 2016 08.
Article in English | MEDLINE | ID: mdl-26861694

ABSTRACT

The initiation of type 1 diabetes (T1D) requires a break in peripheral tolerance. New insights into neoepitope formation indicate that post-translational modification of islet autoantigens, for example via deamidation, may be an important component of disease initiation or exacerbation. Indeed, deamidation of islet autoantigens increases their binding affinity to the T1D highest-risk human leucocyte antigen (HLA) haplotypes HLA-DR3/DQ2 and -DR4/DQ8, increasing the chance that T cells reactive to deamidated autoantigens can be activated upon T cell receptor ligation. Here we investigated human pancreatic islets and inflammatory and tolerogenic human dendritic cells (DC and tolDC) as potential sources of deamidated islet autoantigens and examined whether deamidation is altered in an inflammatory environment. Islets, DC and tolDC contained tissue transglutaminase, the key enzyme responsible for peptide deamidation, and enzyme activity increased following an inflammatory insult. Islets treated with inflammatory cytokines were found to contain deamidated insulin C-peptide. DC, heterozygous for the T1D highest-risk DQ2/8, pulsed with native islet autoantigens could present naturally processed deamidated neoepitopes. HLA-DQ2 or -DQ8 homozygous DC did not present deamidated islet peptides. This study identifies both human islets and DC as sources of deamidated islet autoantigens and implicates inflammatory activation of tissue transglutaminase as a potential mechanism for islet and DC deamidation.


Subject(s)
Amides/chemistry , Autoantigens/immunology , Autoantigens/metabolism , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Protein Processing, Post-Translational , Autoantigens/biosynthesis , Autoantigens/genetics , C-Peptide/immunology , Dendritic Cells/physiology , HLA-DQ Antigens/immunology , HLA-DR3 Antigen/immunology , Humans , Immune Tolerance , Inflammation/immunology , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Proteome , T-Lymphocytes/immunology , Transglutaminases/metabolism
6.
Hepatology ; 62(5): 1536-50, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26185095

ABSTRACT

UNLABELLED: Autoimmune hepatitis (AIH) in humans is a severe inflammatory liver disease characterized by interface hepatitis, the presence of circulating autoantibodies, and hyper-gammaglobulinemia. There are two types of AIH, type 1 (AIH-1) and type 2 (AIH-2), characterized by distinct autoimmune serology. Patients with AIH-1 are positive for anti-smooth muscle and/or antinuclear autoantibodies, whereas patients with AIH-2 have anti-liver kidney microsomal type 1 and/or anti-liver cytosol type 1 autoantibodies. Cytochrome P4502D6 is the antigenic target of anti-liver kidney microsomal type 1, and formiminotransferase cyclodeaminase is the antigenic target of anti-liver cytosol type 1. It is known that AIH, both types 1 and 2, is strongly linked to the human leukocyte antigen (HLA) alleles -DR3, -DR4, and -DR7. However, direct evidence of the association of HLA with AIH is lacking. We developed a novel mouse model of AIH using the HLA-DR3 transgenic mouse on the nonobese-diabetic background by immunization of HLA-DR3- and HLA-DR3+ nonobese-diabetic mice with a DNA plasmid, coding for human cytochrome P4502D6/formiminotransferase cyclodeaminase fusion protein. Immunization with cytochrome P4502D6/formiminotransferase cyclodeaminase leads to a sustained elevation of alanine aminotransferase, development of antinuclear autoantibodies and anti-liver kidney microsomal type 1/anti-liver cytosol type 1 autoantibodies, chronic immune cell infiltration, and parenchymal fibrosis on liver histology in HLA-DR3+ mice. Immunized mice also showed an enhanced T helper 1 immune response and paucity of the frequency of regulatory T cells in the liver. Moreover, HLA-DR3+ mice with exacerbated AIH showed reduced diversity and total load of gut bacteria. CONCLUSION: Our humanized animal model has provided a novel experimental tool to further elucidate the pathogenesis of AIH and to evaluate the efficacy and safety of immunoregulatory therapeutic interventions in vivo.


Subject(s)
Hepatitis, Autoimmune/etiology , Intestines/microbiology , Microbiota , Animals , Autoantibodies/immunology , Base Sequence , Cytochrome P-450 CYP2D6/immunology , Cytokines/biosynthesis , Disease Models, Animal , HLA-DR3 Antigen/immunology , Humans , Immunization , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Molecular Sequence Data , T-Lymphocytes, Regulatory/immunology
7.
J Immunol ; 193(6): 2919-30, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25092888

ABSTRACT

Life-threatening infections caused by Staphylococcus aureus, particularly the community-acquired methicillin-resistant strains of S. aureus, continue to pose serious problems. Greater virulence and increased pathogenicity of certain S. aureus strains are attributed to higher prevalence of exotoxins. Of these exotoxins, the superantigens (SAg) are likely most pathogenic because of their ability to rapidly and robustly activate the T cells even in extremely small quantities. Therefore, countering SAg-mediated T cell activation using T regulatory cells (Tregs) might be beneficial in diseases such as toxic shock syndrome (TSS). As the normal numbers of endogenous Tregs in a typical host are insufficient, we hypothesized that increasing the Treg numbers by administration of IL-2/anti-IL-2 Ab immune complexes (IL2C) or by adoptive transfer of ex vivo expanded Tregs might be more effective in countering SAg-mediated immune activation. HLA-DR3 transgenic mice that closely recapitulate human TSS were treated with IL2C to increase endogenous Tregs or received ex vivo expanded Tregs. Subsequently, they were challenged with SAg to induce TSS. Analyses of various parameters reflective of TSS (serum cytokine/chemokine levels, multiple organ pathology, and SAg-induced peripheral T cell expansion) indicated that increasing the Tregs failed to mitigate TSS. On the contrary, serum IFN-γ levels were increased in IL2C-treated mice. Exploration into the reasons behind the lack of protective effect of Tregs revealed IL-17 and IFN-γ-dependent loss of Tregs during TSS. In addition, significant upregulation of glucocorticoid-induced TNFR family-related receptor on conventional T cells during TSS could render them resistant to Treg-mediated suppression, contributing to failure of Treg-mediated immune regulation.


Subject(s)
Enterotoxins/immunology , Shock, Septic/immunology , Superantigens/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Adoptive Transfer , Animals , Antibodies/immunology , Antibodies/pharmacology , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/pharmacology , Glucocorticoid-Induced TNFR-Related Protein/biosynthesis , Glucocorticoids , HLA-DR alpha-Chains/genetics , HLA-DR alpha-Chains/immunology , HLA-DR beta-Chains/genetics , HLA-DR beta-Chains/immunology , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/immunology , Interferon-gamma/blood , Interferon-gamma/immunology , Interleukin-17/immunology , Interleukin-2/immunology , Interleukin-2/pharmacology , Lymphocyte Activation/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Transgenic , Receptors, Tumor Necrosis Factor/biosynthesis , Shock, Septic/microbiology , Staphylococcal Infections/immunology , Up-Regulation
8.
Am J Gastroenterol ; 110(6): 915-20, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26010309

ABSTRACT

OBJECTIVES: Tissue transglutaminase autoantibodies (tTGAs) represent the first evidence of celiac disease (CD) development. Associations of HLA-DR3-DQA1*05:01-DQB1*02:01 (i.e., DR3-DQ2) and, to a lesser extent, DR4-DQA1*03:01-DQB1*03:02 (i.e., DR4-DQ8) with the risk of CD differ by country, consistent with additional genetic heterogeneity that further refines risk. Therefore, we examined human leukocyte antigen (HLA) factors other than DR3-DQ2 for their contribution to developing tTGAs. METHODS: The Environmental Determinants of Diabetes in the Young (TEDDY) study enrolled 8,676 infants at an increased HLA-DR-DQ risk for type 1 diabetes and CD into a 15-year prospective surveillance follow-up. Of those followed up, 21% (n=1,813) carried DR3-DQ2/DR3-DQ2, 39% (n=3,359) carried DR3-DQ2/DR4-DQ8, 20% (n=1701) carried DR4-DQ8/DR4-DQ8, and 17% (n=1,493) carried DR4-DQ8/DQ4. Within TEDDY, a nested case-control design of 248 children with CD autoimmunity (CDA) and 248 matched control children were genotyped for HLA-B, -DRB3, -DRB4, -DPA1, and -DPB1 genes, and the entire cohort was genotyped for single-nucleotide polymorphisms (SNPs) using the Illumina ImmunoChip. CDA was defined as a positive tTGA test at two consecutive clinic visits, whereas matching in those with no evidence of tTGAs was based on the presence of HLA-DQ2, country, and sex. RESULTS: After adjustment for DR3-DQ2 and restriction to allele frequency (AF) ≥5%, HLA-DPB1*04:01 was inversely associated with CDA by conditional logistic regression (AF=44%, odds ratio=0.71, 95% confidence interval (CI)=0.53-0.96, P=0.025). This association of time to CDA and HLA-DPB1*04:01 was replicated with statistical significance in the remainder of the cohort using imputation for specific HLA alleles based on SNP genotyping (hazard ratio=0.84, 95% CI=0.73-0.96, P=0.013). CONCLUSIONS: HLA-DPB1*04:01 may reduce the risk of tTGAs, an early marker of CD, among DR3-DQ2 children, confirming that additional variants in the HLA region influence the risk for CDA.


Subject(s)
Autoimmune Diseases/genetics , Celiac Disease/genetics , HLA-DP beta-Chains/genetics , Autoantibodies/immunology , Autoimmune Diseases/immunology , Celiac Disease/immunology , Child , Child, Preschool , Cohort Studies , Female , GTP-Binding Proteins/immunology , Gene Frequency , Genetic Predisposition to Disease , HLA-DP beta-Chains/immunology , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/immunology , Humans , Infant , Infant, Newborn , Logistic Models , Male , Polymorphism, Single Nucleotide , Proportional Hazards Models , Protective Factors , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/immunology
9.
Genes Immun ; 15(2): 107-14, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24385024

ABSTRACT

The objective of this study was to study genetic factors that influence quantitative anticyclic citrullinated peptide (anti-CCP) antibody levels in RA patients. We carried out a genome-wide association study (GWAS) meta-analysis using 1975 anti-CCP+ RA patients from three large cohorts, the Brigham Rheumatoid Arthritis Sequential Study (BRASS), North American Rheumatoid Arthritis Consortium (NARAC) and the Epidemiological Investigation of RA (EIRA). We also carried out a genome-wide complex trait analysis (GCTA) to estimate the heritability of anti-CCP levels. GWAS-meta-analysis showed that anti-CCP levels were most strongly associated with the human leukocyte antigen (HLA) region with a P-value of 2 × 10(-11) for rs1980493. There were 112 SNPs in this region that exceeded the genome-wide significance threshold of 5 × 10(-8), and all were in linkage disequilibrium (LD) with the HLA- DRB1*03 allele with LD r(2) in the range of 0.25-0.88. Suggestive novel associations outside of the HLA region were also observed for rs8063248 (near the GP2 gene) with a P-value of 3 × 10(-7). None of the known RA risk alleles (∼52 loci) were associated with anti-CCP level. Heritability analysis estimated that 44% of anti-CCP variation was attributable to genetic factors captured by GWAS variants. In summary, anti-CCP level is a heritable trait, and HLA-DR3 and GP2 are associated with lower anti-CCP levels.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Autoantibodies/immunology , Peptides, Cyclic/immunology , Case-Control Studies , Cohort Studies , Female , GPI-Linked Proteins/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/immunology , HLA-DRB1 Chains/genetics , HLA-DRB1 Chains/immunology , Humans , Linkage Disequilibrium , Male , Middle Aged , Models, Genetic , Polymorphism, Single Nucleotide , Prospective Studies
10.
Infect Immun ; 82(1): 286-97, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166949

ABSTRACT

Unlike human malaria parasites that induce persistent infection, some rodent malaria parasites, like Plasmodium yoelii strain 17XNL (Py17XNL), induce a transient (self-curing) malaria infection. Cooperation between CD4 T cells and B cells to produce antibodies is thought to be critical for clearance of Py17XNL parasites from the blood, with major histocompatibility complex (MHC) class II molecules being required for activation of CD4 T cells. In order to better understand the correspondence between murine malaria models and human malaria, and in particular the role of MHC (HLA) class II molecules, we studied the ability of humanized mice expressing human HLA class II molecules to clear Py17XNL infection. We showed that humanized mice expressing HLA-DR4 (DR0401) molecules and lacking mouse MHC class II molecules (EA(0)) have impaired production of specific antibodies to Py17XNL and cannot cure the infection. In contrast, mice expressing HLA-DR4 (DR0402), HLA-DQ6 (DQ0601), HLA-DQ8 (DQ0302), or HLA-DR3 (DR0301) molecules in an EA(0) background were able to elicit specific antibodies and self-cure the infection. In a series of experiments, we determined that the inability of humanized DR0401.EA(0) mice to elicit specific antibodies was due to expansion and activation of regulatory CD4(+) Foxp3(+) T cells (Tregs) that suppressed B cells to secrete antibodies through cell-cell interactions. Treg depletion allowed the DR0401.EA(0) mice to elicit specific antibodies and self-cure the infection. Our results demonstrated a differential role of MHC (HLA) class II molecules in supporting antibody responses to Py17XNL malaria and revealed a new mechanism by which malaria parasites stimulate B cell-suppressogenic Tregs that prevent clearance of infection.


Subject(s)
B-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , HLA-DR Antigens/immunology , Malaria/immunology , Plasmodium yoelii/immunology , T-Lymphocytes, Regulatory/immunology , Analysis of Variance , Animals , HLA-DQ Antigens/immunology , HLA-DR3 Antigen/immunology , HLA-DR4 Antigen/immunology , Immunity, Cellular/immunology , Immunization , Mice , Mice, Transgenic , T-Lymphocytes, Regulatory/cytology
11.
Clin Immunol ; 152(1-2): 1-9, 2014.
Article in English | MEDLINE | ID: mdl-24576620

ABSTRACT

This study was undertaken to test the hypothesis that Sjogren's syndrome Antigen A (SSA)/Ro60-reactive T cells are activated by peptides originating from oral and gut bacteria. T cell hybridomas generated from HLA-DR3 transgenic mice recognized 3 regions on Ro60, with core epitopes mapped to amino acids 228-238, 246-256 and 371-381. BLAST analysis identified several mimicry peptides, originating from human oral, intestinal, skin and vaginal bacteria, as well as environmental bacteria. Amongst these, a peptide from the von Willebrand factor type A domain protein (vWFA) from the oral microbe Capnocytophaga ochracea was the most potent activator. Further, Ro60-reactive T cells were activated by recombinant vWFA protein and whole Escherichia coli expressing this protein. These results demonstrate that peptides derived from normal human microbiota can activate Ro60-reactive T cells. Thus, immune responses to commensal microbiota and opportunistic pathogens should be explored as potential triggers for initiating autoimmunity in SLE and Sjögren's syndrome.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Lupus Erythematosus, Systemic/immunology , Molecular Mimicry/immunology , Ribonucleoproteins/immunology , Sjogren's Syndrome/immunology , Amino Acid Sequence , Animals , Autoimmunity/immunology , Capnocytophaga/genetics , Capnocytophaga/immunology , Cross Reactions/immunology , Female , HLA-DR3 Antigen/immunology , Humans , Hybridomas/immunology , Intestines/microbiology , Lymphocyte Activation/immunology , Mice , Mouth/microbiology , Peptides/genetics , Peptides/immunology , Recombinant Proteins/immunology , Skin/microbiology , T-Lymphocytes/immunology , Vagina/microbiology , von Willebrand Factor/genetics , von Willebrand Factor/immunology
12.
Clin Exp Immunol ; 178(2): 342-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24962673

ABSTRACT

Sarcoidosis is a granulomatous disorder of unknown aetiology. The presence of Mycobacterium tuberculosis catalase-peroxidase (mKatG) in sarcoidosis tissue has been reported. T helper type 1 (Th1) responses against mKatG have previously been observed. However, little is known about interleukin (IL)-17 and Th17 responses in sarcoidosis. Here, we investigated the levels of IL-17 and frequencies of IL-17-producing cells responding to mKatG in sarcoidosis patients with different prognosis. Peripheral blood and bronchoalveolar lavage (BAL) cells were obtained from sarcoidosis patients with or without Löfgren's syndrome (often associated with spontaneous recovery), and also stratified according to human leucocyte antigen (HLA) type. Cells producing IL-17 and interferon (IFN)-γ after stimulation with mKatG were enumerated by enzyme-linked immunospot (ELISPOT). The level of IL-17 in the BAL fluid of sarcoidosis patients and healthy controls was measured by quantitative immuno-polymerase chain reaction (qIPCR). We also performed flow cytometry and immunohistochemistry for further characterization of IL-17 expression. Patients with Löfgren's syndrome had a higher frequency of IL-17-producing cells responding to mKatG in BAL fluid compared to patients without Löfgren's syndrome (P < 0·05). The HLA-DR3(+) sarcoidosis patients with Löfgren's syndrome (known to have a particularly good prognosis) also had a clearly higher level of IL-17 in BAL fluid compared to healthy controls and sarcoidosis patients without Löfgren's syndrome (P < 0·01) and (P < 0·05), respectively. No such difference between patient groups was observed with regard to IFN-γ and not with regard to either cytokine in peripheral blood. These findings suggest that IL-17-producing cells may be a useful biomarker for the prognosis of sarcoidosis and play a role in the spontaneous recovery typical of patients with Löfgren's syndrome.


Subject(s)
Interleukin-17/metabolism , Sarcoidosis, Pulmonary/immunology , Sarcoidosis, Pulmonary/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Adult , Bacterial Proteins/immunology , Bronchoalveolar Lavage Fluid/immunology , Catalase/immunology , Female , Gene Expression , HLA-DR3 Antigen/immunology , Humans , Interleukin-17/genetics , Lymphocyte Activation/immunology , Male , Middle Aged , Risk Factors , Sarcoidosis, Pulmonary/diagnosis , Sarcoidosis, Pulmonary/genetics , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
13.
Clin Exp Immunol ; 172(1): 16-22, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23480181

ABSTRACT

Interleukin (IL)-21 and protein tyrosine phosphatase non-receptor 22 (PTPN22) regulate lymphocyte function and have been implicated in the pathogenesis of autoimmune diabetes. We sequenced the proximal promoter of the IL-21 gene for the first time and analysed the PTPN22 1858T polymorphism in type 1A diabetes (T1AD) patients and healthy controls (HC). We correlated the frequencies of islet and extra-pancreatic autoantibodies with genotypes from both loci. The case series comprised 612 T1AD patients and 792 HC. Genotyping of PTPN22 C1858T was performed on 434 T1AD patients and 689 HC. The -448 to +83 base pairs (bp) region of the IL-21 gene was sequenced in 309 Brazilian T1AD and 189 HC subjects. We also evaluated human leucocyte antigen (HLA) DR3/DR4 alleles. The frequencies of glutamic acid decarboxylase (GAD65), tyrosine phosphatase-like protein (IA)-2, anti-nuclear antibody (ANA), thyroid peroxidase (TPO), thyroglobulin (TG), thyrotrophin receptor autoantibody (TRAb), anti-smooth muscle (ASM) and 21-hydroxylase (21-OH) autoantibodies were higher in T1AD patients than in HC. The PTPN22 1858T allele was associated with an increased risk for developing T1AD [odds ratio (OR) = 1·94; P < 0·001], particularly in patients of European ancestry, and with a higher frequency of GAD65 and TG autoantibodies. HLA-DR3/DR4 alleles predominated in T1AD patients. A heterozygous allelic IL-21 gene variant (g.-241 T > A) was found in only one patient. In conclusion, only PTPN22 C1858T polymorphism and HLA-DR3 and/or DR4 alleles, but not allelic variants in the 5'-proximal region of the IL-21 gene were associated with T1AD risk. Patients with T1AD had increased frequencies of anti-islet-cell, anti-thyroid, anti-nuclear, anti-smooth muscle and anti-21-OH autoantibodies. The C1858T PTPN22 polymorphism was also associated with a higher frequency of GAD65 and TG autoantibodies.


Subject(s)
Autoantibodies/immunology , Diabetes Mellitus, Type 1/genetics , Interleukins/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Alleles , Autoantibodies/genetics , Autoantibodies/metabolism , Biomarkers/metabolism , Brazil , Case-Control Studies , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Female , Gene Expression , Gene Frequency , Genetic Predisposition to Disease , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/immunology , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/immunology , HLA-DR4 Antigen/genetics , HLA-DR4 Antigen/immunology , Humans , Interleukins/immunology , Male , Promoter Regions, Genetic , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Risk , Sequence Analysis, DNA , White People
14.
J Immunol ; 186(10): 6056-63, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21471440

ABSTRACT

Recently we demonstrated that zinc transporter 8 (ZnT8) is a major target of autoantibodies in human type 1 diabetes (T1D). Because the molecules recognized by T1D autoantibodies are typically also targets of autoreactive T cells, we reasoned that this would likely be the case for ZnT8. To test this hypothesis, IFN-γ-producing T cells specific for ZnT8 in the peripheral blood of 35 patients with T1D (<6 mo after onset at blood draw) and 41 age-matched controls were assayed by ELISPOT using a library of 23 overlapping dipeptide pools covering the entire 369 aa primary sequence. Consistent with our hypothesis, patients showed significantly higher T cell reactivity than the matched controls, manifest in terms of the breadth of the overall response and the magnitude of responses to individual pools. Therefore, the median number of pools giving positive responses (stimulation index ≥ 3) in the control group was 1.0 (range, 0-7) compared with 6.0 (range, 1-20; p < 0.0001) for the patients. Similarly, the median stimulation index of positive responses in controls was 3.1 versus 5.0 in the patients (p < 0.0001). Individually, 7 of 23 pools showed significant disease association (p < 0.001), with several of the component peptides binding the disease associated HLA-DR3 (0301) and -DR4 (0401) molecules in vitro. We conclude that ZnT8 is also a major target of disease-associated autoreactive T cells in human T1D, and we suggest that reagents that target ZnT8-specific T cells could have therapeutic potential in preventing or arresting the progression of this disease.


Subject(s)
Autoantibodies/blood , Autoantibodies/immunology , Cation Transport Proteins/immunology , Diabetes Mellitus, Type 1/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Autoimmunity , Child , Enzyme-Linked Immunospot Assay , Female , Genotype , HLA-DR3 Antigen/immunology , HLA-DR4 Antigen/immunology , Humans , Interferon-gamma/biosynthesis , Male , Zinc Transporter 8
15.
J Biol Chem ; 286(11): 9737-47, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21233204

ABSTRACT

T-cell stimulating activity of Staphylococcal enterotoxin B (SEB) is an important factor in the pathogenesis of certain staphylococcal diseases including SEB mediated shock. SEB is one of the most potent superantigens known and treatment of SEB induced shock remains a challenge. We generated and characterized murine monoclonal antibodies (mAbs) to SEB in mice. We tested mAbs neutralize mitogenic effects of SEB in vitro and in vivo with T-cell proliferation assays and 2 murine models for SEB induced lethal shock (SEBILS). Epitope mapping suggests that all these mAbs recognize conformational epitopes that are destroyed by deleting the C terminus of the protein. Further site-directed mutagenesis identified potential residues involved in binding to SEB that differ between Methicillin resistant and sensitive Staphylococcus aureus strains. Only mAb 20B1 was effective as a monotherapy in treating SEBILS in HLA DR3 transgenic mice, which exhibit enhanced sensitivity to SEB. It is noteworthy that mAbs, 14G8 and 6D3 were not protective when given alone in the HLA DR3 mice but their efficacy of protection could be greatly enhanced when mAbs were co-administered simultaneously. Our data suggest combinations of defined mAbs may constitute a better treatment strategy and provide a new insight for the development of passive immunotherapy.


Subject(s)
Antibodies, Bacterial/pharmacology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Neutralizing/pharmacology , Enterotoxins/toxicity , Methicillin-Resistant Staphylococcus aureus/immunology , Shock/prevention & control , Animals , Antibodies, Bacterial/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neutralizing/immunology , Enterotoxins/genetics , Enterotoxins/immunology , Epitope Mapping/methods , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/immunology , Methicillin Resistance/immunology , Methicillin-Resistant Staphylococcus aureus/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mutagenesis, Site-Directed , Shock/chemically induced , Shock/immunology
16.
Tissue Antigens ; 80(4): 356-62, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22835336

ABSTRACT

Microsatellite polymorphism in exon 5 of major histocompatibility complex class I chain related gene-A (MIC-A) has been implicated in the etiology of autoimmune diseases including type 1 diabetes (T1D) and celiac disease (CD). In this study on North Indian population, the MIC-A5.1 allele, carrying a premature termination codon in transmembrane region, was observed with increased frequency in T1D (29.6%, odds ratio OR = 2.1, P = 0.00017) and CD patients (40.3%, OR = 3.37, P = 1.67E-05) than in controls (16.7%). When the MIC-A5.1 association was adjusted for linkage with human leukocyte antigen (HLA)-DR3, the statistical significance of the association was abolished. This implies that the observed association of MIC-A5.1 is due to its linkage disequilibrium (D' = 0.94) with HLA-B8-DR3-DQ2 haplotype and is secondary to the overall association with DR3 positive MHC haplotypes.


Subject(s)
Celiac Disease/genetics , Diabetes Mellitus, Type 1/genetics , Histocompatibility Antigens Class I/genetics , Polymorphism, Genetic , Adolescent , Adult , Alleles , Case-Control Studies , Celiac Disease/immunology , Child , Child, Preschool , Diabetes Mellitus, Type 1/immunology , Female , Gene Frequency , HLA-B8 Antigen/genetics , HLA-B8 Antigen/immunology , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/immunology , Haplotypes , Histocompatibility Antigens Class I/immunology , Humans , India , Linkage Disequilibrium , Male , Middle Aged , Protein Isoforms/genetics , Protein Isoforms/immunology , White People
17.
Am J Pathol ; 178(6): 2760-73, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21641398

ABSTRACT

Among the exotoxins produced by Staphylococcus aureus and Streptococcus pyogenes, the superantigens (SAgs) are the most potent T-cell activators known to date. SAgs are implicated in several serious diseases including toxic shock syndrome (TSS), Kawasaki disease, and sepsis. However, the immunopathogenesis of TSS and other diseases involving SAgs are still not completely understood. The commonly used conventional laboratory mouse strains do not respond robustly to SAgs in vivo. Therefore, they must be artificially rendered susceptible to TSS by using sensitizing agents such as d-galactosamine (d-galN), which skews the disease exclusively to the liver and, hence, is not representative of the disease in humans. SAg-induced TSS was characterized using transgenic mice expressing HLA class II molecules that are extremely susceptible to TSS without d-galN. HLA-DR3 transgenic mice recapitulated TSS in humans with extensive multiple-organ inflammation affecting the lung, liver, kidneys, heart, and small intestines. Heavy infiltration with T lymphocytes (both CD4(+) and CD8+), neutrophils, and macrophages was noted. In particular, the pathologic changes in the small intestines were extensive and accompanied by significantly altered absorptive functions of the enterocytes. In contrast to massive liver failure alone in the d-galN sensitization model of TSS, findings of the present study suggest that gut dysfunction might be a key pathogenic event that leads to high morbidity and mortality in humans with TSS.


Subject(s)
HLA-DR3 Antigen/immunology , Liver/immunology , Liver/pathology , Shock, Septic/immunology , Shock, Septic/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Disease Models, Animal , Enterotoxins/immunology , Galactosamine/immunology , Gastrointestinal Tract/pathology , Humans , Imaging, Three-Dimensional , Immunization , Mice , Mice, Transgenic , Organ Specificity/immunology , Permeability , Shock, Septic/blood , Shock, Septic/etiology , Time Factors
18.
J Autoimmun ; 38(4): 344-53, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22459490

ABSTRACT

Previously we showed that transgenic mice expressing human HLA-DR3 gene are susceptible to PLP(91-110) induced experimental autoimmune encephalomyelitis (EAE) and can serve as an animal model of multiple sclerosis (MS). HLA-DR3 mice with EAE showed increased number of CD8 T cells indicating their important role in disease pathogenesis. The role of CD8 T cells in MS, an inflammatory demyelinating disease of CNS, has been enigmatic as it has been assigned both regulatory and pathogenic roles. Therefore, to evaluate the role of CD8 T cells, we generated CD8 deficient HLA-DR3 transgenic mice (DR3.CD8(-/-)). Immunization with PLP(91-110) led to more severe EAE in DR3.CD8(-/-) mice compared to HLA-DR3 mice indicating a regulatory role for CD8 T cells. Interestingly, DR3.CD8(-/-) mice with EAE showed decreased CNS pathology compared to DR3 mice thus suggesting a pathogenic role for CD8 T cells. We show that these two subsets of CD8 T cells can be differentiated based on the surface expression of CD122 (IL-2 Rß chain). CD8 T cells expressing CD122 (CD8+CD122+) play a regulatory role while CD8+CD122- T cells act as a pathogenic subset. CD122 expressing CD8 T cells are the regulatory subset of CD8 T cells and regulate the encephalitogenic CD4 T cells through direct modulation of antigen presenting cells and/or through the release of immunoregulatory cytokines such as IL-10, IFNγ and TGFß. We also showed that adoptive transfer of CD8CD122- T cells caused increased spinal cord demyelination indicating that these are pathogenic subset of CD8 T cells. Our study suggests that CD8+ T cells play both regulatory as well as pathogenic role in disease pathogenesis of EAE. A better understanding of these subsets could aid in designing novel therapy for MS patients.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , HLA-DR3 Antigen/genetics , Myelin Proteolipid Protein/immunology , Adoptive Transfer , Animals , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Central Nervous System/immunology , Central Nervous System/pathology , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic , Demyelinating Diseases/genetics , Demyelinating Diseases/immunology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , HLA-DR3 Antigen/immunology , Interleukin-2 Receptor beta Subunit/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteolipid Protein/adverse effects , Myelin Proteolipid Protein/chemistry , Neuroglia/immunology
19.
J Immunol ; 184(2): 1085-91, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20007529

ABSTRACT

Anti-Smith (Sm) D autoantibodies are specific for systemic lupus erythematosus. In this investigation, the influence of HLA-D genes on immune responses to SmD was investigated. Mice with HLA-DR3, HLA-DR4, HLA-DQ0601, HLA-DQ0604, or HLA-DQ8 transgenes were immunized with recombinant SmD1, and their Ab responses were analyzed. Analysis by ELISA showed that all strains responded well to SmD. However, when synthetic SmD peptides were used as substrate, DR3 mice had the highest Ab response followed by DQ8, DQ0604, DQ0601, and DR4. A similar trend was observed in Western blot analysis using WEHI 7.1 cell lysate as the substrate, with the exception that DR4 mice did not generate detectable amounts of Abs. Only sera from DR3 and DQ0604 mice immunoprecipitated A-ribonucleoprotein (RNP), SmB, and SmD. Intermolecular epitope spreading to A-RNP and SmB was evident in DR3 and DQ0604 mice, as sera depleted of anti-SmD Abs were reactive with these proteins. DR3 mice also generated an immune response to C-RNP. Anti-nuclear Abs were detected in the majority of the DR3 mice, whereas moderate reactivities were seen in DQ0604 and DQ8 mice. Interestingly, only DR3 mice mounted an anti-dsDNA Ab response. Approximately half of the anti-dsDNA Abs were cross-reactive with SmD. Ab responses correlated with the strength of the T cell responses. Thus, HLA-DR3 appears to be the dominant HLA-D gene that determines the magnitude and quality of the anti-SmD immune response. In addition, our findings provide insights into the origin of the anti-dsDNA Abs often detected in patients with systemic lupus erythematosus.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , HLA-DQ Antigens/immunology , HLA-DR Antigens/immunology , Lupus Erythematosus, Systemic/immunology , Animals , Antibody Diversity , Antigens, Nuclear/immunology , Autoantibodies/biosynthesis , Autoimmunity , DNA/immunology , HLA-DQ Antigens/genetics , HLA-DR Antigens/genetics , HLA-DR3 Antigen/immunology , Mice , Mice, Transgenic , Ribonucleoproteins, Small Nuclear/immunology , snRNP Core Proteins
20.
J Autoimmun ; 37(3): 254-62, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21868195

ABSTRACT

Although systemic lupus erythematosus (SLE) is a multigenic autoimmune disorder, HLA-D is the most dominant genetic susceptibility locus. This study was undertaken to investigate the hypothesis that microbial peptides bind HLA-DR3 and activate T cells reactive with lupus autoantigens. Using HLA-DR3 transgenic mice and lupus-associated autoantigen SmD protein, SmD(79-93) was identified to contain a dominant HLA-DR3 restricted T cell epitope. This T cell epitope was characterized by using a T-T hybridoma, C1P2, generated from SmD immunized HLA-DR3 transgenic mouse. By pattern search analysis, 20 putative mimicry peptides (P2-P21) of SmD(79-93,) from microbial and human origin were identified. C1P2 cells responded to SmD, SmD(79-93) and a peptide (P20) from Vibro cholerae. Immunization of HLA-DR3 mice with P20 induced T cell responses and IgG antibodies to SmD that were not cross-reactive with the immunogen. A T-T hybridoma, P20P1, generated from P20 immunized mice, not only responded to P20 and SmD(79-93), but also to peptides from Streptococcus agalactiae (P17) and human-La related protein (P11). These three T cell mimics (P20, P11 and P17) induced diverse and different autoantibody response profiles. Our data demonstrates for the first time molecular mimicry at T cell epitope level between lupus-associated autoantigen SmD and microbial peptides. Considering that distinct autoreactive T cell clones were activated by different microbial peptides, molecular mimicry at T cell epitope level can be an important pathway for the activation of autoreactive T cells resulting in the production of autoantibodies. In addition, the novel findings reported herein may have significant implications in the pathogenesis of SLE.


Subject(s)
Autoantibodies/immunology , Autoimmunity , HLA-DR3 Antigen/immunology , Lupus Erythematosus, Systemic/immunology , Peptides/immunology , T-Lymphocytes/immunology , snRNP Core Proteins/immunology , Animals , Antibody Formation/drug effects , Autoantibodies/metabolism , Autoantigens/immunology , Autoantigens/metabolism , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , HLA-DR3 Antigen/metabolism , Humans , Hybridomas/immunology , Hybridomas/metabolism , Immunization , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Molecular Mimicry , Protein Binding , Streptococcus agalactiae/immunology , Streptococcus agalactiae/metabolism , T-Lymphocytes/metabolism , Vibrio cholerae/immunology , Vibrio cholerae/metabolism , snRNP Core Proteins/administration & dosage , snRNP Core Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL