Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Lupus ; 21(10): 1113-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22472776

ABSTRACT

OBJECTIVES: Homozygous C1q deficiency is an extremely rare condition and strongly associated with systemic lupus erythematosus. To assess and characterize C1q deficiency in an African-American lupus pedigree, C1q genomic region was evaluated in the lupus cases and family members. METHODS: Genomic DNA from patient was obtained and C1q A, B and C gene cluster was sequenced using next generation sequencing method. The identified mutation was further confirmed by direct Sanger sequencing method in the patient and all blood relatives. C1q levels in serum were measured using sandwich ELISA method. RESULTS: In an African-American patient with lupus and C1q deficiency, we identified and confirmed a novel homozygote start codon mutation in C1qA gene that changes amino acid methionine to arginine at position 1. The Met1Arg mutation prevents protein translation (Met1Arg). Mutation analyses of the patient's family members also revealed the Met1Arg homozygote mutation in her deceased brother who also had lupus with absence of total complement activity consistent with a recessive pattern of inheritance. CONCLUSION: The identification of new mutation in C1qA gene that disrupts the start codon (ATG to AGG (Met1Arg)) has not been reported previously and it expands the knowledge and importance of the C1q gene in the pathogenesis of lupus especially in the high-risk African-American population.


Subject(s)
Complement C1q/deficiency , Complement C1q/genetics , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/genetics , Point Mutation , Adult , Black or African American/genetics , Amino Acid Substitution , Base Sequence , Codon, Initiator/genetics , DNA Mutational Analysis , Female , Genes, Recessive , Homozygote , Humans , Male , Pedigree , Young Adult
2.
Genes Immun ; 13(5): 380-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22476155

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease with diverse clinical manifestations characterized by the development of pathogenic autoantibodies manifesting in inflammation of target organs such as the kidneys, skin and joints. Genome-wide association studies have identified genetic variants in the UBE2L3 region that are associated with SLE in subjects of European and Asian ancestry. UBE2L3 encodes an ubiquitin-conjugating enzyme, UBCH7, involved in cell proliferation and immune function. In this study, we sought to further characterize the genetic association in the region of UBE2L3 and use molecular methods to determine the functional effect of the risk haplotype. We identified significant associations between variants in the region of UBE2L3 and SLE in individuals of European and Asian ancestry that exceeded a Bonferroni-corrected threshold (P<1 × 10(-4)). A single risk haplotype was observed in all associated populations. Individuals harboring the risk haplotype display a significant increase in both UBE2L3 mRNA expression (P=0.0004) and UBCH7 protein expression (P=0.0068). The results suggest that variants carried on the SLE-associated UBE2L3 risk haplotype influence autoimmunity by modulating UBCH7 expression.


Subject(s)
Genetic Predisposition to Disease , Haplotypes , Lupus Erythematosus, Systemic/genetics , Ubiquitin-Conjugating Enzymes/genetics , Black or African American/genetics , Alleles , Asian People/genetics , Female , Hispanic or Latino/genetics , Humans , Linkage Disequilibrium , Lupus Erythematosus, Systemic/ethnology , Male , Polymorphism, Single Nucleotide , Ubiquitin-Conjugating Enzymes/metabolism , White People/genetics
3.
Clin Exp Immunol ; 159(3): 281-91, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19968664

ABSTRACT

Interferon (IFN) signature genes have been shown to be expressed highly in peripheral blood of patients with systemic lupus erythematosus (SLE), especially in the presence of active disease. However, the expression of this gene signature in individuals with incomplete forms of lupus and the pathogenic relationship between IFN signature genes and autoantibody production have not been explored fully. In the present study, we examined the gene expression and autoantibody profiles of patients diagnosed with incomplete lupus erythematosus (ILE) to determine correlations of the gene expression signature with autoantibody production. Gene expression analysis was carried out on the 24K Illumina Human Refseq-8 arrays using blood samples from 84 subjects, including patients with SLE (n = 27) or ILE (n = 24), first-degree relatives (FDR) of these patients (n = 22) and non-autoimmune control (NC) individuals (n = 11). Autoantibody expression was measured using standard immunoassays and autoantigen proteomic arrays. Up-regulation of a set of 63 IFN signature genes was seen in 83% of SLE patients and 50% of ILE patients. High levels of IFN gene expression in ILE and SLE showed significant correlations with the expression of a subset of IgG autoantibodies, including chromatin, dsDNA, dsRNA, U1snRNP, Ro/SSA, La/SSB, topoisomerase I and Scl 70, while low IFN levels were correlated with immunoglobulin (Ig)M autoreactivity. These studies suggest that in patients with ILE the IFN gene expression signature may identify a subset of these individuals who are at risk for disease progression. Furthermore, high levels of alpha IFN may promote autoantibody class-switch from IgM to the more pathogenic IgG class.


Subject(s)
Autoantibodies/biosynthesis , Gene Expression Regulation , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Interferons/metabolism , Lupus Erythematosus, Systemic/metabolism , Adult , Antibody Formation/genetics , Antibody Formation/immunology , Autoantibodies/genetics , Autoantibodies/immunology , Autoantigens/chemistry , Autoantigens/immunology , Female , Gene Expression Profiling/methods , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Interferons/genetics , Interferons/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Proteomics/methods , Somatic Hypermutation, Immunoglobulin/genetics , Somatic Hypermutation, Immunoglobulin/immunology
4.
Genes Immun ; 10(5): 517-24, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19440201

ABSTRACT

Complement cascade plasma proteins play a complex role in the etiopathogenesis of systemic lupus erythematosus (SLE). Hereditary C1q deficiency has been strongly related to SLE; however, there are very few published SLE studies that evaluate the polymorphisms of genes encoding for C1q (A, B and C). In this study, we evaluated 17 single nucleotide polymorphisms (SNPs) across 37 kb of C1QA, C1QB and C1QC in a lupus cohort of individuals of the African-American and Hispanic origin. In a case-only analysis, a significant association at multiple SNPs in the C1QA gene was detected in African Americans with kidney nephritis (best P=4.91 x 10(-6)). In addition, C1QA was associated with SLE in African Americans with a lack of nephritis and accompanying photosensitivity when compared with that in normal controls (P=6.80 x 10(-6)). A similar trend was observed in the Hispanic subjects (P=0.003). Quantitative analysis showed that some SNPs in C1q genes might be correlated with C3 complement levels in an additive model among African Americans (best P=0.0001). The C1QA gene is associated with subphenotypes of lupus in the African-American and Hispanic subjects. Further studies with higher SNP densities in this region and other complement components are necessary to elucidate the complex genetics and phenotypic interactions between complement components and SLE.


Subject(s)
Complement C1q/genetics , Genetic Predisposition to Disease , Lupus Erythematosus, Systemic/ethnology , Lupus Erythematosus, Systemic/genetics , Polymorphism, Single Nucleotide , Black or African American/genetics , Hispanic or Latino/genetics , Humans , Lupus Erythematosus, Systemic/physiopathology , Lupus Nephritis/genetics , Oklahoma/ethnology
5.
Genes Immun ; 10(5): 531-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19339986

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease with highly variable clinical presentation. Patients suffer from immunological abnormalities that target T-cell, B-cell and accessory cell functions. B cells are hyperactive in SLE patients. An adapter protein expressed in B cells called BANK1 (B-cell scaffold protein with ankyrin repeats) was reported in a previous study to be associated with SLE in a European population. The objective of this study was to assess the BANK1 genotype-phenotype association in an independent replication sample. We genotyped 38 single nucleotide polymorphisms (SNPs) in BANK1 on 1892 European-derived SLE patients and 2652 European-derived controls. The strongest associations with SLE and BANK1 were at rs17266594 (corrected P-value=1.97 x 10(-5), odds ratio (OR)=1.22, 95% CI 1.12-1.34) and rs10516487 (corrected P-value=2.59 x 10(-5), OR=1.22, 95% CI 1.11-1.34). Our findings suggest that the association is explained by these two SNPs, confirming previous reports that these polymorphisms contribute to the risk of developing lupus. Analysis of patient subsets enriched for hematological, immunological and renal ACR criteria or the levels of autoantibodies, such as anti-RNP A and anti-SmRNP, uncovers additional BANK1 associations. Our results suggest that BANK1 polymorphisms alter immune system development and function to increase the risk for developing lupus.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Genetic Predisposition to Disease , Lupus Erythematosus, Systemic/genetics , Membrane Proteins/genetics , Polymorphism, Single Nucleotide , Adaptor Proteins, Signal Transducing/immunology , Case-Control Studies , Humans , Lupus Erythematosus, Systemic/immunology , Membrane Proteins/immunology , White People/genetics
6.
Genes Immun ; 10(5): 397-403, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19369946

ABSTRACT

We targeted LYN, a src-tyosine kinase involved in B-cell activation, in case-control association studies using populations of European-American, African-American and Korean subjects. Our combined European-derived population, consisting of 2463 independent cases and 3131 unrelated controls, shows significant association with rs6983130 in a female-only analysis with 2254 cases and 2228 controls (P=1.1 x 10(-4), odds ratio (OR)=0.81 (95% confidence interval: 0.73-0.90)). This single nucleotide polymorphism (SNP) is located in the 5' untranslated region within the first intron near the transcription initiation site of LYN. In addition, SNPs upstream of the first exon also show weak and sporadic association in subsets of the total European-American population. Multivariate logistic regression analysis implicates rs6983130 as a protective factor for systemic lupus erythematosus (SLE) susceptibility when anti-dsDNA, anti-chromatin, anti-52 kDa Ro or anti-Sm autoantibody status were used as covariates. Subset analysis of the European-American female cases by American College of Rheumatology classification criteria shows a reduction in the risk of hematological disorder with rs6983130 compared with cases without hematological disorders (P=1.5 x 10(-3), OR=0.75 (95% CI: 0.62-0.89)). None of the 90 SNPs tested show significant association with SLE in the African American or Korean populations. These results support an association of LYN with European-derived individuals with SLE, especially within autoantibody or clinical subsets.


Subject(s)
Lupus Erythematosus, Systemic/genetics , Polymorphism, Single Nucleotide , src-Family Kinases/genetics , Age Factors , Case-Control Studies , Female , Genome-Wide Association Study , Humans , Lupus Erythematosus, Systemic/epidemiology , Lupus Erythematosus, Systemic/immunology
7.
Genes Immun ; 10(5): 503-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19262577

ABSTRACT

Sle3 is a NZM2410/NZW-derived lupus-susceptibility interval on murine chromosome 7, which is associated with spontaneous lupus nephritis (SLN), and also anti-GBM-induced glomerulonephritis (GN). The tissue kallikrein gene cluster is located within the Sle3 interval and constitutes potential candidate genes for this locus. We have recently reported that renal kallikrein expression was upregulated by anti-GBM antibody challenge in a strain-specific manner and that it was significantly underexpressed in the anti-GBM-sensitive strains, including B6.Sle3. Further sequencing and functional studies reported earlier provided evidence that kallikreins could constitute disease genes in lupus. In this report, we have used an adenoviral vector to deliver the klk1 gene to B6.Sle3 congenics to directly test if kallikreins might have a protective effect against anti-GBM-induced nephritis. Our data show that klk1 gene delivery ameliorated anti-GBM-induced nephritis in B6.Sle3 congenics. Taken together with earlier studies, these findings indicate that kallikreins play an important protective role in autoantibody-initiated GN and could constitute potential candidate genes for anti-GBM-induced GN and SLN.


Subject(s)
Autoantibodies/immunology , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Tissue Kallikreins/genetics , Animals , Mice , Mice, Congenic , Mice, Inbred C57BL , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Tissue Kallikreins/immunology
8.
Lupus ; 17(12): 1070-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19029274

ABSTRACT

Individuals suffering from systemic lupus erythematosus (SLE) are predisposed to accelerate cardiovascular disease. Our laboratory has recently developed an animal model of SLE-accelerated atherosclerosis. We have shown that, following 8 weeks feeding high fat Western diet, radiation chimeras consisting of SLE-derived haematopoietic cells transferred to low-density lipoprotein (LDL)r(-/-) mice (LDLr.Sle) have increased atherosclerosis compared with C57Bl/6 bone marrow recipients (LDLr.B6). However, this feeding regimen resulted in significant mortality in SLE-susceptible mice compared with controls with surviving animals having extremely elevated serum cholesterol (>500 mg/dL) and increased serum markers of kidney pathology. To test the hypothesis that SLE-associated autoimmune dysregulation can exacerbate atherosclerosis under more mild serum cholesterol conditions (approximately 200 mg/dL), we examined SLE and lesion development in radiation chimeras fed either a normal chow or high fat Western diet for 8 weeks. High fat fed LDLr.Sle mice exhibited increased mortality and were significantly more hypertensive. LDLr.Sle mice had greater titres of antibodies against dsDNA, oxLDL and phospholipid compared with controls. Lupus-susceptibility increased the atherosclerotic lesions and the percentage of CD4(+) T cells in the lesions of proximal aortas, independent of diet. These data show that increased dyslipidemia resulting from high-fat feeding can exacerbate autoimmunity and associated vascular complications. Conversely, they also show that autoimmune dysregulation can accelerate atherosclerosis in LDLr-deficient animals independent of feeding high fat diet. Collectively this study provides additional evidence that the accelerated atherosclerosis observed in SLE is autoimmune associated.


Subject(s)
Atherosclerosis , Dietary Fats/pharmacology , Lupus Erythematosus, Systemic , Receptors, LDL/genetics , Animals , Antibodies, Antiphospholipid/metabolism , Atherosclerosis/complications , Atherosclerosis/genetics , Atherosclerosis/immunology , Autoimmunity/immunology , Cholesterol, LDL/metabolism , Cholesterol, VLDL/metabolism , DNA/immunology , Dyslipidemias/complications , Dyslipidemias/genetics , Dyslipidemias/immunology , Lipoproteins, LDL/metabolism , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Receptors, LDL/metabolism
9.
Genes Immun ; 9(3): 187-94, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18288123

ABSTRACT

Increased expression of interferon (IFN)-inducible genes is implicated in the pathogenesis of systemic lupus erythematosus (SLE). One transcription factor responsible for regulating IFN, interferon regulatory factor-5 (IRF5), has been associated with SLE in genetic studies of Asian, Caucasian and Hispanic populations. We genotyped up to seven polymorphic loci in or near IRF5 in a total of 4870 African-American and Caucasian subjects (1829 SLE sporadic cases and 3041 controls) from two independent studies. Population-based case-control comparisons were performed using the Pearson's chi(2)-test statistics and haplotypes were inferred using HaploView. We observed significant novel associations with the IRF5 variants rs2004640 and rs3807306 in African Americans and replicated previously reported associations in Caucasians. While we identified risk haplotypes, the majority of haplotypic effects were accounted for by one SNP (rs3807306) in conditional analyses. We conclude that genetic variants of IRF5 associate with SLE in multiple populations, providing evidence that IRF5 is likely to be a crucial component in SLE pathogenesis among multiple ethnic groups.


Subject(s)
Black or African American/genetics , Interferon Regulatory Factors/genetics , Lupus Erythematosus, Systemic/genetics , Gene Frequency , Genetics, Population , Genotype , Haplotypes/genetics , Humans , Interferon Regulatory Factors/metabolism , Linkage Disequilibrium , Polymorphism, Single Nucleotide
10.
Genes Immun ; 9(1): 61-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18094711

ABSTRACT

The evolutionary origin of genetic diversity in the SLAM/CD2 gene cluster, implicated in autoimmune lupus susceptibility in mice, was investigated by sequence analysis of exons from six members of the cluster in 48 wild mouse samples derived from the global mouse population. A total of 80 coding region SNPs were identified among the six genes analyzed, indicating that this gene cluster is highly polymorphic in natural mouse populations. Phylogenetic analyses of these allelic sequences revealed clustering of alleles derived from multiple Mus species and subspecies, indicating alleles at several SLAM/CD2 loci were present in ancestral Mus populations prior to speciation and have persisted as polymorphisms for more than 1 million years. Analyses of nonsynonymous/synonymous ratios using likelihood codon substitution models identified several segments in Cd229, Cd48 and Cd84 that were impacted by positive diversifying selective pressures. These findings support the interpretation that selection favoring the generation and retention of functional polymorphisms has played a role in the evolutionary origin of genetic polymorphisms that are predisposing to autoimmunity.


Subject(s)
Alleles , Autoimmunity/genetics , Evolution, Molecular , Genetic Predisposition to Disease , Genetics, Population , Amino Acid Sequence , Animals , Animals, Laboratory , Animals, Wild , Antigens, CD/genetics , Antigens, CD/immunology , Autoimmunity/immunology , Codon , Exons , Haplotypes , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Molecular Sequence Data , Multigene Family , Phylogeny , Polymorphism, Single Nucleotide , Prevalence , Protein Structure, Tertiary , Selection, Genetic , Sequence Analysis, DNA , Sequence Homology, Amino Acid
11.
Ann Rheum Dis ; 67(4): 458-61, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17720724

ABSTRACT

OBJECTIVE: The aetiology of systemic lupus erythematosus (SLE) is incompletely understood. Both genetic and environmental factors are implicated in the pathogenesis of the disease. Herein, we describe genetic association between SLE and polymorphisms in the interleukin (IL)-21 gene. The reported effect of IL-21 on B-cell differentiation into plasma cells and its effect on dendritic cell maturation and T-cell responses make IL-21 an attractive candidate gene for SLE. METHODS: Three single nucleotide polymorphisms (SNPs) in the IL-21 gene were genotyped in a total of 2636 individuals (1318 cases and 1318 controls matched for age, sex and race). Population-based case-control association analyses were performed. RESULTS: We found a genetic association with SLE and two SNPs located within the IL-21 gene (rs907715: chi(2) = 11.55, p<0.001; rs2221903: chi(2) = 5.49, p = 0.019). Furthermore, genotypes homozygous for the risk alleles were more frequent than genotypes homozygous for the non-risk alleles in European-American patients as compared to controls (rs907715 (GG versus AA): odds ratio (OR) = 1.66, p = 0.0049; rs2221903 (GG versus AA): OR = 1.60, p = 0.025). CONCLUSION: Our findings indicate that IL-21 polymorphism is a candidate association with SLE. The functional effects of this association, when revealed, might improve our understanding of the disease and provide new therapeutic targets.


Subject(s)
Interleukins/genetics , Lupus Erythematosus, Systemic/genetics , Polymorphism, Single Nucleotide , Black or African American/genetics , Case-Control Studies , Female , Gene Frequency , Genetic Predisposition to Disease , Genotype , Haplotypes , Humans , Lupus Erythematosus, Systemic/ethnology , Male , White People/genetics
12.
Genes Immun ; 8(8): 634-45, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17728789

ABSTRACT

Genetic analyses of the lupus-prone NZM2410 mouse have identified multiple susceptibility loci on chromosome 7, termed Sle3 and Sle5. Both of these loci were contained within a large congenic interval, originally termed as Sle3 that strongly impacts a variety of myeloid and T-cell phenotypes and mediates fatal lupus nephritis when combined with Sle1. We have now produced two subcongenic strains, B6.Sle3 and B6.Sle5, carrying the Sle3 and Sle5 intervals separately and characterized their phenotypes as monocongenic strains and individually in combination with Sle1. Neither B6.Sle3 nor B6.Sle5 monocongenic strain develop severe autoimmunity; however, both of these intervals cause the development of severe glomerulonephritis when combined with Sle1. Thus, B6.Sle1Sle3 and B6.Sle1Sle5 exhibit splenomegaly, expansion of activated B and CD4+ T-cell populations and high levels of IgG and IgM autoantibodies targeting multiple nuclear antigens, intact glomeruli and various other autoantigens. In addition, B6.Sle1Sle3 mice also produced higher levels of IgA antinuclear autoantibodies, which were implicated in the development of IgA nephropathy. Our results indicate that Sle3 and Sle5 can independently complement with Sle1, through shared and unique mechanisms, to mediate the development of severe autoimmunity.


Subject(s)
Autoimmunity/genetics , Genetic Testing , Lupus Nephritis/genetics , Animals , Autoantigens/analysis , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chromosomes/genetics , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Lupus Nephritis/pathology , Lymphocyte Activation/genetics , Mice , Mice, Congenic , Protein Array Analysis , Splenomegaly/immunology
13.
Genes Immun ; 8(7): 604-12, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17713556

ABSTRACT

The inhibitory receptor FcgammaRIIb regulates B-cell functions. Genetic studies have associated Fcgr2b polymorphisms and lupus susceptibility in both humans and murine models, in which B cells express reduced FcgammaRIIb levels. Furthermore, FcgammaRIIb absence results in lupus on the appropriate genetic background, and lentiviral-mediated FcgammaRIIb overexpression prevents disease in the NZM2410 lupus mouse. The NZM2410/NZW allele Fcgr2b is, however, located in-between Sle1a and Sle1b, two potent susceptibility loci, making it difficult to evaluate Fcr2b(NZW) independent contribution. By using two congenic strains that each carries only Sle1a (B6.Sle1a(15-353)), or Fcr2b(NZW) in the absence of Sle1a or Sle1b (B6.Sle1(111-148)), we show that the Fcr2b(NZW) allele does not upregulate its expression on germinal center B cells and plasma cells, as does the C57BL/6 allele on B6.Sle1a(15-353) B cells. Furthermore, in the absence of the flanking Sle1a and Sle1b, Fcr2b(NZW) does not produce an autoimmune phenotype, but is associated with an increased number of class-switched plasma cells. These results show that while a lower level of FcgammaRIIb does not by itself induce the development of autoreactive B cells, it has the potential to amplify the contribution of autoreactive B cells induced by other lupus-susceptibility loci by enhancing the production of class-switched plasma cells.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Immunoglobulin G/biosynthesis , Receptors, IgG/genetics , Alleles , Animals , B-Lymphocytes/cytology , Dendritic Cells, Follicular/cytology , Dendritic Cells, Follicular/immunology , Gene Expression Regulation , Genetic Predisposition to Disease , Germinal Center/cytology , Germinal Center/metabolism , Immunoglobulin Class Switching , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred Strains , Plasma Cells/immunology , Receptors, IgG/immunology , Receptors, IgG/metabolism , Up-Regulation
14.
Clin Exp Immunol ; 147(1): 60-70, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17177964

ABSTRACT

The objective of this study was to investigate the prevalence and clinical significance of a spectrum of autoantibodies in systemic lupus erythematosus and incomplete lupus syndromes using a proteome microarray bearing 70 autoantigens. Microarrays containing candidate autoantigens or control proteins were printed on 16-section slides. These arrays were used to profile 93 serum samples from patients with systemic lupus erythematosus (SLE (n = 33), incomplete LE (ILE; n = 23), first-degree relatives (FDRs) of SLE patients (n = 20) and non-autoimmune controls (NC; n = 17). Data were analysed using the significance analysis of microarray (SAM) and clustering algorithms. Correlations with disease features were determined. Serum from ILE and SLE patients contained high levels of IgG autoantibodies to 50 autoantigens and IgM autoantibodies to 12 autoantigens. Elevated levels of at least one IgG autoantibody were detected in 26% of SLE and 19% of ILE samples; elevated IgM autoantibodies were present in 13% of SLE and 17% of ILE samples. IgG autoantibodies segregated into seven clusters including two specific for DNA and RNA autoantigens that were correlated with the number of lupus criteria. Three IgG autoantibody clusters specific for collagens, DNA and histones, were correlated with renal involvement. Of the four IgM autoantibody clusters, two were correlated negatively with the number of lupus criteria; none were correlated with renal disease. The IgG : IgM autoantibody ratios generally showed a stepwise increase in the groups following disease burden from NC to SLE. Insights derived from the expanded autoantibody profiling made possible with the antigen array suggest differences in autoreactivity in ILE and SLE. Determining whether the IgM aurotreactivity that predominates in ILE represents an early stage prior to IgG switching or is persistent and relatively protective will require further longitudinal studies.


Subject(s)
Autoantibodies/blood , Data Interpretation, Statistical , Lupus Erythematosus, Systemic/immunology , Protein Array Analysis/methods , Adult , Analysis of Variance , Autoantigens/isolation & purification , Case-Control Studies , Female , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , In Situ Hybridization , Lupus Erythematosus, Systemic/genetics , Lupus Nephritis/immunology , Male , Middle Aged
15.
Lupus ; 14(8): 598-606, 2005.
Article in English | MEDLINE | ID: mdl-16175931

ABSTRACT

Systemic lupus erythematosus (SLE) is characterized by loss of immune tolerance. A hallmark of SLE is the presence of autoantibodies resulting from B cell hyperactivity. Previous studies have shown that the presence of abnormal B cell subsets in the periphery, such as CD27highCD20- B cells, correlate with disease activity. We examined the relationship between the expression of CD70, the ligand for CD27 expressed by activated T cells, and indicators of disease activity. A significant increase in median CD70+CD4+ T cell frequencies and memory CD45RA-CD4+ T cell frequencies was observed in SLE samples as compared to healthy controls. The frequencies of CD70+CD4+ T cells correlated with disease duration but not age, treatment, or disease activity. Although a majority of CD70+CD4+ T cells appeared to be effector memory cells, mitogen-stimulated CD70+CD4+ T cells were capable of secreting a full repertoire of effector cytokines. Despite the presence of activated CD4+ T cells, no increase in immunosenescent CD4+ T cells, as defined by the loss of CD28 and/or the acquisition of CD57 was observed in samples from SLE patients. These studies indicate that increased CD70 expression might serve as a useful marker of abnormal T cell activity in SLE.


Subject(s)
Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/metabolism , Lupus Erythematosus, Systemic/immunology , Membrane Proteins/metabolism , T-Lymphocyte Subsets/metabolism , Tumor Necrosis Factors/metabolism , Adult , CD27 Ligand , CD4 Lymphocyte Count , Case-Control Studies , Female , Humans , Leukocyte Common Antigens/metabolism , Lupus Erythematosus, Systemic/blood , Lymphocyte Activation , Middle Aged
16.
Lupus ; 14(1): 2-12, 2005.
Article in English | MEDLINE | ID: mdl-15732281

ABSTRACT

Susceptibility to lupus nephritis is the end-result of complex interactions between polymorphic genetic factors involved in the regulation of immune responses. In humans, genome-wide screens and candidate-gene analyses led to the identification of several loci containing potential targets (FcgammaRIIa, PTPN22, PD-1, IL-10) for physiopathological research and therapeutic interventions. In mice, the generation of congenic mice, bearing in a normal genetic background one single disease-associated locus, greatly improved our understanding of the mechanisms mediating the genetic contribution to the disease. In the future, the identification of disease-associated genes will open new perspectives for the development of more targeted therapies of lupus nephritis.


Subject(s)
Genetic Predisposition to Disease/genetics , Lupus Nephritis/genetics , Animals , Genetic Testing , Humans , Lupus Nephritis/immunology , Mice , Models, Animal
17.
Immunogenetics ; 56(5): 333-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15309345

ABSTRACT

Genetic control of homeostasis of peripheral CD4+ lymphocyte levels is incompletely understood. Recent genome scans have linked mouse peripheral CD4 levels to chromosome 17, with strongest linkage to the Ea region. Nonobese diabetic (NOD) mice demonstrate peripheral T-cell lymphocytosis, and previous studies also suggested that the MHC region might control this phenotype. Here we confirm that loci on Chr 17 control NOD peripheral CD4 lymphocytosis. An elevated NOD CD4:CD8 ratio maps to the same region, and we show it is due to increased numbers of CD4+ cells. However, using NOD MHC congenic mice, we demonstrate that the MHC region is excluded, and that NOD peripheral lymphocytosis is controlled by genetic intervals adjacent to the MHC region on Chr 17.


Subject(s)
CD4-Positive T-Lymphocytes , Lymphocytosis/genetics , Animals , Chromosome Mapping , Genetic Markers , Lymphocytosis/etiology , Mice , Mice, Inbred NOD
18.
Genes Immun ; 3(3): 144-50, 2002 May.
Article in English | MEDLINE | ID: mdl-12070778

ABSTRACT

The ratio of CD4 T cells to CD8 T cells (CD4:CD8 ratio) varies over two-fold between C57BL/6 and DBA/2 mice for both T cell precursors in the thymus and mature T cells in the periphery. Correlation analysis of the CD4:CD8 ratio in thymic precursors vs peripheral T cells in F2 and backcross mice found that thymic precursor ratios are inherited independently from those in the periphery, indicating that the CD4:CD8 ratios in these populations are affected by distinct genetic mechanisms. A genome scan of progeny in the phenotypic extremes identified three quantitative trait loci (QTLs). Trmq1 (for T cell ratio modifier QTL 1) was detected in the telomeric end of c6 (peak marker D6Mit15 at 74 cM) and had a maximum LOD score of 4.6. Trmq2, in the telomeric half of c2, peaked at D2MIT483 and had a maximum LOD score of 3.41. Both of these QTLs impacted the CD4:CD8 ratios in peripheral T cells and had no impact on variation in this ratio among thymic precursors. However, heterozygosity for the H2 complex was suggestively associated (LOD score of 2.43) with increases in CD4 T cells among T cell precursors in the thymus. All of these QTLs were affected by epistatic interactions, indicating that additional modifiers in the B6 and DBA/2 genomes modulate this phenotype.


Subject(s)
CD4-CD8 Ratio , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Genetic Linkage , Animals , Chromosome Mapping , Crosses, Genetic , Lod Score , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Species Specificity , Thymus Gland/metabolism
19.
Immunity ; 15(5): 775-85, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11728339

ABSTRACT

The major murine systemic lupus erythematosus (SLE) susceptibility locus, Sle1, corresponds to three loci independently affecting loss of tolerance to chromatin in the NZM2410 mouse. The congenic interval corresponding to Sle1c contains Cr2, which encodes complement receptors 1 and 2 (CR1/CR2, CD35/CD21). NZM2410/NZW Cr2 exhibits a single nucleotide polymorphism that introduces a novel glycosylation site, resulting in higher molecular weight proteins. This polymorphism, located in the C3d binding domain, reduces ligand binding and receptor-mediated cell signaling. Molecular modeling based on the recently solved CR2 structure in complex with C3d reveals that this glycosylation interferes with receptor dimerization. These data demonstrate a functionally significant phenotype for the NZM2410 Cr2 allele and strongly support its role as a lupus susceptibility gene.


Subject(s)
Lupus Erythematosus, Systemic/genetics , Receptors, Complement 3d/genetics , Alleles , Animals , Genetic Predisposition to Disease , Lupus Erythematosus, Systemic/immunology , Mice , Polymorphism, Genetic , Receptors, Complement 3d/immunology
20.
Immunity ; 15(3): 397-408, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11567630

ABSTRACT

Genetic predisposition plays a crucial role in susceptibility to systemic lupus erythematosus (SLE) in both human patients and animal models. Recent progress in experimental systems and human linkage analysis is providing key insights into the genetic basis for susceptibility and elucidating the manner in which genetic interactions mediate severe disease pathogenesis. Genes in multiple pathways appear to participate in specific elements of the disease, and epistatic interactions among these genes play an important role in both aggravating and suppressing disease development.


Subject(s)
Lupus Erythematosus, Systemic/genetics , Autoimmunity , Chromosome Mapping , Complement C4/genetics , Genetic Linkage , HLA Antigens/genetics , Humans , Receptors, IgG/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...