Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Am J Hum Genet ; 107(5): 864-881, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33031749

ABSTRACT

Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disorder with a prominent genetic component. Individuals of African ancestry (AA) experience the disease more severely and with an increased co-morbidity burden compared to European ancestry (EA) populations. We hypothesize that the disparities in disease prevalence, activity, and response to standard medications between AA and EA populations is partially conferred by genomic influences on biological pathways. To address this, we applied a comprehensive approach to identify all genes predicted from SNP-associated risk loci detected with the Immunochip. By combining genes predicted via eQTL analysis, as well as those predicted from base-pair changes in intergenic enhancer sites, coding-region variants, and SNP-gene proximity, we were able to identify 1,731 potential ancestry-specific and trans-ancestry genetic drivers of SLE. Gene associations were linked to upstream and downstream regulators using connectivity mapping, and predicted biological pathways were mined for candidate drug targets. Examination of trans-ancestral pathways reflect the well-defined role for interferons in SLE and revealed pathways associated with tissue repair and remodeling. EA-dominant genetic drivers were more often associated with innate immune and myeloid cell function pathways, whereas AA-dominant pathways mirror clinical findings in AA subjects, suggesting disease progression is driven by aberrant B cell activity accompanied by ER stress and metabolic dysfunction. Finally, potential ancestry-specific and non-specific drug candidates were identified. The integration of all SLE SNP-predicted genes into functional pathways revealed critical molecular pathways representative of each population, underscoring the influence of ancestry on disease mechanism and also providing key insight for therapeutic selection.


Subject(s)
Gene Regulatory Networks , Genome, Human , Interferons/genetics , Lupus Erythematosus, Systemic/ethnology , Lupus Erythematosus, Systemic/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Black People , Bortezomib/therapeutic use , DNA, Intergenic/genetics , DNA, Intergenic/immunology , Enhancer Elements, Genetic , Gene Expression , Gene Ontology , Genetic Predisposition to Disease , Genome-Wide Association Study , Heterocyclic Compounds/therapeutic use , Humans , Interferons/immunology , Isoquinolines/therapeutic use , Lactams , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Molecular Sequence Annotation , Protein Array Analysis , Quantitative Trait, Heritable , White People
2.
Front Immunol ; 14: 1147526, 2023.
Article in English | MEDLINE | ID: mdl-36936908

ABSTRACT

Introduction: Pathologic inflammation is a major driver of kidney damage in lupus nephritis (LN), but the immune mechanisms of disease progression and risk factors for end organ damage are poorly understood. Methods: To characterize molecular profiles through the development of LN, we carried out gene expression analysis of microdissected kidneys from lupus-prone NZM2328 mice. We examined male mice and the congenic NZM2328.R27 strain as a means to define mechanisms associated with resistance to chronic nephritis. Gene expression profiles in lupus mice were compared with those in human LN. Results: NZM2328 mice exhibited progress from acute to transitional and then to chronic glomerulonephritis (GN). Each stage manifested a unique molecular profile. Neither male mice nor R27 mice progressed past the acute GN stage, with the former exhibiting minimal immune infiltration and the latter enrichment of immunoregulatory gene signatures in conjunction with robust kidney tubule cell profiles indicative of resistance to cellular damage. The gene expression profiles of human LN were similar to those noted in the NZM2328 mouse suggesting comparable stages of LN progression. Conclusions: Overall, this work provides a comprehensive examination of the immune processes involved in progression of murine LN and thus contributes to our understanding of the risk factors for end-stage renal disease. In addition, this work presents a foundation for improved classification of LN and illustrates the applicability of murine models to identify the stages of human disease.


Subject(s)
Glomerulonephritis , Kidney Failure, Chronic , Lupus Nephritis , Humans , Mice , Male , Animals , Kidney/pathology , Glomerulonephritis/pathology , Inflammation , Kidney Failure, Chronic/pathology , Chronic Disease
3.
Sci Rep ; 11(1): 14789, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34285256

ABSTRACT

To compare lupus pathogenesis in disparate tissues, we analyzed gene expression profiles of human discoid lupus erythematosus (DLE) and lupus nephritis (LN). We found common increases in myeloid cell-defining gene sets and decreases in genes controlling glucose and lipid metabolism in lupus-affected skin and kidney. Regression models in DLE indicated increased glycolysis was correlated with keratinocyte, endothelial, and inflammatory cell transcripts, and decreased tricarboxylic (TCA) cycle genes were correlated with the keratinocyte signature. In LN, regression models demonstrated decreased glycolysis and TCA cycle genes were correlated with increased endothelial or decreased kidney cell transcripts, respectively. Less severe glomerular LN exhibited similar alterations in metabolism and tissue cell transcripts before monocyte/myeloid cell infiltration in some patients. Additionally, changes to mitochondrial and peroxisomal transcripts were associated with specific cells rather than global signal changes. Examination of murine LN gene expression demonstrated metabolic changes were not driven by acute exposure to type I interferon and could be restored after immunosuppression. Finally, expression of HAVCR1, a tubule damage marker, was negatively correlated with the TCA cycle signature in LN models. These results indicate that altered metabolic dysfunction is a common, reversible change in lupus-affected tissues and appears to reflect damage downstream of immunologic processes.


Subject(s)
Gene Expression Profiling/methods , Gene Regulatory Networks , Lupus Erythematosus, Discoid/genetics , Lupus Nephritis/genetics , Animals , Citric Acid Cycle , Databases, Genetic , Disease Models, Animal , Female , Gene Expression Regulation , Glucose/metabolism , Glycolysis , Humans , Interferon Type I/adverse effects , Lipid Metabolism , Lupus Erythematosus, Discoid/metabolism , Lupus Nephritis/metabolism , Mice
4.
Genes (Basel) ; 12(12)2021 11 26.
Article in English | MEDLINE | ID: mdl-34946847

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic, multisystem, autoimmune inflammatory disease with genomic and non-genomic contributions to risk. We hypothesize that epigenetic factors are a significant contributor to SLE risk and may be informative for identifying pathogenic mechanisms and therapeutic targets. To test this hypothesis while controlling for genetic background, we performed an epigenome-wide analysis of DNA methylation in genomic DNA from whole blood in three pairs of female monozygotic (MZ) twins of European ancestry, discordant for SLE. Results were replicated on the same array in four cell types from a set of four Danish female MZ twin pairs discordant for SLE. Genes implicated by the epigenetic analyses were then evaluated in 10 independent SLE gene expression datasets from the Gene Expression Omnibus (GEO). There were 59 differentially methylated loci between unaffected and affected MZ twins in whole blood, including 11 novel loci. All but two of these loci were hypomethylated in the SLE twins relative to the unaffected twins. The genes harboring these hypomethylated loci exhibited increased expression in multiple independent datasets of SLE patients. This pattern was largely consistent regardless of disease activity, cell type, or renal tissue type. The genes proximal to CpGs exhibiting differential methylation (DM) in the SLE-discordant MZ twins and exhibiting differential expression (DE) in independent SLE GEO cohorts (DM-DE genes) clustered into two pathways: the nucleic acid-sensing pathway and the type I interferon pathway. The DM-DE genes were also informatically queried for potential gene-drug interactions, yielding a list of 41 drugs including a known SLE therapy. The DM-DE genes delineate two important biologic pathways that are not only reflective of the heterogeneity of SLE but may also correlate with distinct IFN responses that depend on the source, type, and location of nucleic acid molecules and the activated receptors in individual patients. Cell- and tissue-specific analyses will be critical to the understanding of genetic factors dysregulating the nucleic acid-sensing and IFN pathways and whether these factors could be appropriate targets for therapeutic intervention.


Subject(s)
DNA Methylation/genetics , Diseases in Twins/genetics , Interferons/genetics , Lupus Erythematosus, Systemic/genetics , Nucleic Acids/genetics , Signal Transduction/genetics , Twins, Monozygotic/genetics , DNA/genetics , Drug Delivery Systems/methods , Epigenomics/methods , Female , Genetic Techniques , Humans , Promoter Regions, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL