Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
Genome Med ; 15(1): 84, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37845772

ABSTRACT

BACKGROUND: Systemic lupus erythematosus (SLE) is known to be clinically heterogeneous. Previous efforts to characterize subsets of SLE patients based on gene expression analysis have not been reproduced because of small sample sizes or technical problems. The aim of this study was to develop a robust patient stratification system using gene expression profiling to characterize individual lupus patients. METHODS: We employed gene set variation analysis (GSVA) of informative gene modules to identify molecular endotypes of SLE patients, machine learning (ML) to classify individual patients into molecular subsets, and logistic regression to develop a composite metric estimating the scope of immunologic perturbations. SHapley Additive ExPlanations (SHAP) revealed the impact of specific features on patient sub-setting. RESULTS: Using five datasets comprising 2183 patients, eight SLE endotypes were identified. Expanded analysis of 3166 samples in 17 datasets revealed that each endotype had unique gene enrichment patterns, but not all endotypes were observed in all datasets. ML algorithms trained on 2183 patients and tested on 983 patients not used to develop the model demonstrated effective classification into one of eight endotypes. SHAP indicated a unique array of features influential in sorting individual samples into each of the endotypes. A composite molecular score was calculated for each patient and significantly correlated with standard laboratory measures. Significant differences in clinical characteristics were associated with different endotypes, with those with the least perturbed transcriptional profile manifesting lower disease severity. The more abnormal endotypes were significantly more likely to experience a severe flare over the subsequent 52 weeks while on standard-of-care medication and specific endotypes were more likely to be clinical responders to the investigational product tested in one clinical trial analyzed (tabalumab). CONCLUSIONS: Transcriptomic profiling and ML reproducibly separated lupus patients into molecular endotypes with significant differences in clinical features, outcomes, and responsiveness to therapy. Our classification approach using a composite scoring system based on underlying molecular abnormalities has both staging and prognostic relevance.


Subject(s)
Lupus Erythematosus, Systemic , Transcriptome , Humans , Gene Expression Profiling , Gene Regulatory Networks , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/drug therapy , Algorithms
3.
Ann Rheum Dis ; 81(5): 632-643, 2022 05.
Article in English | MEDLINE | ID: mdl-35115332

ABSTRACT

OBJECTIVES: The goals of these studies were to elucidate the inter-relationships of specific anti-nuclear antibody (ANA), complement, and the interferon gene signature (IGS) in the pathogenesis of systemic lupus erythematosus (SLE). METHODS: Data from the Illuminate trials were analysed for antibodies to dsDNA as well as RNA-binding proteins (RBP), levels of C3, C4 and various IGS. Statistical hypothesis testing, linear regression analyses and classification and regression trees analysis were employed to assess relationships between the laboratory features of SLE. RESULTS: Inter-relationships of ANAs, complement and the IGS differed between patients of African Ancestry (AA) and European Ancestry (EA); anti-RNP and multiple autoantibodies were more common in AA patients and, although both related to the presence of the IGS, relationships between autoantibodies and complement differed. Whereas, anti-dsDNA had an inverse relationship to C3 and C4, levels of anti-RNP were not related to these markers. The IGS was only correlated with anti-dsDNA in EA SLE and complement was more correlated to the IGS in AA SLE. Finally, autoantibodies occurred in the presence and absence of the IGS, whereas the IGS was infrequent in anti-dsDNA/anti-RBP-negative SLE patients. CONCLUSION: There is a complex relationship between autoantibodies and the IGS, with anti-RNP associated in AA and both anti-dsDNA and RNP associated in EA. Moreover, there was a difference in the relationship between anti-dsDNA, but not anti-RBP, with complement levels. The lack of a relationship of anti-RNP with C3 and C4 suggests that anti-RNP immune complexes (ICs) may drive the IGS without complement fixation, whereas anti-dsDNA ICs involve complement consumption.


Subject(s)
Complement C3 , Complement C4 , Interferons , Lupus Erythematosus, Systemic , Antibodies, Antinuclear/immunology , Antigen-Antibody Complex , Antiviral Agents , Autoantibodies/immunology , Complement C3/immunology , Complement C3/metabolism , Complement C4/immunology , Complement C4/metabolism , DNA , Humans , Interferons/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism
4.
Curr Opin Rheumatol ; 33(6): 579-585, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34410228

ABSTRACT

PURPOSE OF REVIEW: To summarize recent studies stratifying SLE patients into subgroups based on gene expression profiling and suggest future improvements for employing transcriptomic data to foster precision medicine. RECENT FINDINGS: Bioinformatic & machine learning pipelines have been employed to dissect the transcriptomic heterogeneity of lupus patients and identify more homogenous subgroups. Some examples include the use of unsupervised random forest and k-means clustering to separate adult SLE patients into seven clusters and hierarchical clustering of single-cell RNA-sequencing (scRNA-seq) of immune cells yielding four clusters in a cohort of adult SLE and pediatric SLE participants. Random forest classification of bulk RNA-seq data from sorted blood cells enabled prediction of high or low disease activity in European and Asian SLE patients. Inferred transcription factor activity stratified adult and pediatric SLE into two subgroups. SUMMARY: Several different endotypes of SLE patients with differing molecular profiles have been reported but a global consensus of clinically actionable groups has not been reached. Moreover, heterogeneity between datasets, reproducibility of predictions as well as the most effective classification approach have not been resolved. Nevertheless, gene expression-based precision medicine remains an attractive option to subset lupus patients.


Subject(s)
Lupus Erythematosus, Systemic , Precision Medicine , Gene Expression Profiling , Humans , Lupus Erythematosus, Systemic/genetics , Reproducibility of Results , Transcriptome
5.
Sci Rep ; 11(1): 7052, 2021 03 29.
Article in English | MEDLINE | ID: mdl-33782412

ABSTRACT

SARS-CoV2 is a previously uncharacterized coronavirus and causative agent of the COVID-19 pandemic. The host response to SARS-CoV2 has not yet been fully delineated, hampering a precise approach to therapy. To address this, we carried out a comprehensive analysis of gene expression data from the blood, lung, and airway of COVID-19 patients. Our results indicate that COVID-19 pathogenesis is driven by populations of myeloid-lineage cells with highly inflammatory but distinct transcriptional signatures in each compartment. The relative absence of cytotoxic cells in the lung suggests a model in which delayed clearance of the virus may permit exaggerated myeloid cell activation that contributes to disease pathogenesis by the production of inflammatory mediators. The gene expression profiles also identify potential therapeutic targets that could be modified with available drugs. The data suggest that transcriptomic profiling can provide an understanding of the pathogenesis of COVID-19 in individual patients.


Subject(s)
Bronchi/metabolism , COVID-19/metabolism , Gene Expression Profiling , Lung/metabolism , Bronchoalveolar Lavage Fluid , COVID-19/blood , COVID-19/virology , Humans , Inflammation Mediators/metabolism , Myeloid Cells/metabolism , Protein Binding , SARS-CoV-2/isolation & purification
6.
Sci Rep ; 10(1): 17361, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33060686

ABSTRACT

Arthritis is a common manifestation of systemic lupus erythematosus (SLE) yet understanding of the underlying pathogenic mechanisms remains incomplete. We, therefore, interrogated gene expression profiles of SLE synovium to gain insight into the nature of lupus arthritis (LA), using osteoarthritis (OA) and rheumatoid arthritis (RA) as comparators. Knee synovia from SLE, OA, and RA patients were analyzed for differentially expressed genes (DEGs) and also by Weighted Gene Co-expression Network Analysis (WGCNA) to identify modules of highly co-expressed genes. Genes upregulated and/or co-expressed in LA revealed numerous immune/inflammatory cells dominated by a myeloid phenotype, in which pathogenic macrophages, myeloid-lineage cells, and their secreted products perpetuate inflammation, whereas OA was characterized by fibroblasts and RA of lymphocytes. Genes governing trafficking of immune cells into the synovium by chemokines were identified, but not in situ generation of germinal centers (GCs). Gene Set Variation Analysis (GSVA) confirmed activation of specific immune cell types in LA. Numerous therapies were predicted to target LA, including TNF, NFκB, MAPK, and CDK inhibitors. Detailed gene expression analysis identified a unique pattern of cellular components and physiologic pathways operative in LA, as well as drugs potentially able to target this common manifestation of SLE.


Subject(s)
Arthritis/genetics , Gene Expression Profiling , Lupus Erythematosus, Systemic/genetics , Myeloid Cells/pathology , Arthritis/pathology , Down-Regulation , Humans , Lupus Erythematosus, Systemic/pathology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...