Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 388(3): 751-764, 2024 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-37673681

RESUMEN

Toll-like receptor 7 (TLR7) and TLR8 are single-stranded RNA-sensing endosomal pattern recognition receptors that evolved to defend against viral infections. However, aberrant TLR7/8 activation by endogenous ligands has been implicated in the pathogenesis of autoimmune diseases including systemic lupus erythematosus. TLR activation and type I interferon (IFN) were shown recently to impart resistance to glucocorticoids (GC), which are part of the standard of care for multiple autoimmune diseases. While GCs are effective, a plethora of undesirable effects limit their use. New treatment approaches that allow for the use of lower and safer doses of GCs would be highly beneficial. Herein, we report that a dual TLR7/8 inhibitor (TLR7/8i) increases the effectiveness of GCs in inflammatory settings. Human peripheral blood mononuclear cell studies revealed increased GC sensitivity in the presence of TLR7/8i for reducing inflammatory cytokine production, a synergistic effect that was most pronounced in myeloid cells, particularly monocytes. Gene expression analysis by NanoString and single-cell RNA sequencing revealed that myeloid cells were substantially impacted by combining low-dose TLR7/8i and GC, as evidenced by the effects on nuclear factor-kappa B-regulated cytokines and GC-response genes, although IFNs were affected to a smaller degree. Low dose of TLR7/8i plus GC was more efficacious then either agent alone in the MRL/lpr mouse model of lupus, with improved proteinuria and survival. Overall, our findings indicate a GC-sparing potential for TLR7/8i compounds, suggesting TLR7/8i may offer a new strategy for the treatment of autoimmune diseases. SIGNIFICANCE STATEMENT: Some features of autoimmune diseases may be resistant to glucocorticoids, mediated at least in part by toll-like receptor (TLR) activation, necessitating higher doses that are associated with considerable toxicities. We demonstrate that TLR7/8 inhibition and glucocorticoids work synergistically to reduce inflammation in a cell-type specific manner and suppress disease in a mouse model of lupus. TLR7/8 inhibition is a promising strategy for the treatment of autoimmune diseases and has glucocorticoid-sparing potential.


Asunto(s)
Lupus Eritematoso Sistémico , Receptor Toll-Like 7 , Ratones , Animales , Humanos , Receptor Toll-Like 7/metabolismo , Glucocorticoides/farmacología , Leucocitos Mononucleares/metabolismo , Ratones Endogámicos MRL lpr , Receptores Toll-Like , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/genética
3.
Genome Med ; 15(1): 84, 2023 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-37845772

RESUMEN

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.


Asunto(s)
Lupus Eritematoso Sistémico , Transcriptoma , Humanos , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/tratamiento farmacológico , Algoritmos
4.
Genes (Basel) ; 12(12)2021 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-34946847

RESUMEN

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.


Asunto(s)
Metilación de ADN/genética , Enfermedades en Gemelos/genética , Interferones/genética , Lupus Eritematoso Sistémico/genética , Ácidos Nucleicos/genética , Transducción de Señal/genética , Gemelos Monocigóticos/genética , ADN/genética , Sistemas de Liberación de Medicamentos/métodos , Epigenómica/métodos , Femenino , Técnicas Genéticas , Humanos , Regiones Promotoras Genéticas/genética
5.
Sci Rep ; 11(1): 14789, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34285256

RESUMEN

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.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Lupus Eritematoso Discoide/genética , Nefritis Lúpica/genética , Animales , Ciclo del Ácido Cítrico , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Glucosa/metabolismo , Glucólisis , Humanos , Interferón Tipo I/efectos adversos , Metabolismo de los Lípidos , Lupus Eritematoso Discoide/metabolismo , Nefritis Lúpica/metabolismo , Ratones
6.
Front Immunol ; 12: 649458, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815408

RESUMEN

A distinct B cell population marked by elevated CD11c expression is found in patients with systemic lupus erythematosus (SLE). Cells with a similar phenotype have been described during chronic infection, but variable gating strategies and nomenclature have led to uncertainty of their relationship to each other. We isolated CD11chi cells from peripheral blood and characterized them using transcriptome and IgH repertoire analyses. Gene expression data revealed the CD11chi IgD+ and IgD- subsets were highly similar to each other, but distinct from naive, memory, and plasma cell subsets. Although CD11chi B cells were enriched in some germinal center (GC) transcripts and expressed numerous negative regulators of B cell receptor (BCR) activation, they were distinct from GC B cells. Gene expression patterns from SLE CD11chi B cells were shared with other human diseases, but not with mouse age-associated B cells. IgH V-gene sequencing analysis showed IgD+ and IgD- CD11chi B cells had somatic hypermutation and were clonally related to each other and to conventional memory and plasma cells. However, the IgH repertoires expressed by the different subsets suggested that defects in negative selection during GC transit could contribute to autoimmunity. The results portray a pervasive B cell population that accumulates during autoimmunity and chronic infection and is refractory to BCR signaling.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Cadenas Pesadas de Inmunoglobulina/genética , Infecciones/inmunología , Lupus Eritematoso Sistémico/inmunología , Adulto , Anciano , Animales , Subgrupos de Linfocitos B/metabolismo , Antígeno CD11c/metabolismo , Biología Computacional , Conjuntos de Datos como Asunto , Femenino , Perfilación de la Expresión Génica , Centro Germinal/citología , Humanos , Cadenas Pesadas de Inmunoglobulina/metabolismo , Infecciones/sangre , Lupus Eritematoso Sistémico/sangre , Ratones , Persona de Mediana Edad
7.
Am J Hum Genet ; 107(5): 864-881, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33031749

RESUMEN

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.


Asunto(s)
Redes Reguladoras de Genes , Genoma Humano , Interferones/genética , Lupus Eritematoso Sistémico/etnología , Lupus Eritematoso Sistémico/genética , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Linfocitos B/inmunología , Linfocitos B/patología , Población Negra , Bortezomib/uso terapéutico , ADN Intergénico/genética , ADN Intergénico/inmunología , Elementos de Facilitación Genéticos , Expresión Génica , Ontología de Genes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Compuestos Heterocíclicos/uso terapéutico , Humanos , Interferones/inmunología , Isoquinolinas/uso terapéutico , Lactamas , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Anotación de Secuencia Molecular , Análisis por Matrices de Proteínas , Carácter Cuantitativo Heredable , Población Blanca
8.
Sci Rep ; 10(1): 17361, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33060686

RESUMEN

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.


Asunto(s)
Artritis/genética , Perfilación de la Expresión Génica , Lupus Eritematoso Sistémico/genética , Células Mieloides/patología , Artritis/patología , Regulación hacia Abajo , Humanos , Lupus Eritematoso Sistémico/patología , Regulación hacia Arriba
9.
JCI Insight ; 5(15)2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32759501

RESUMEN

Gene expression signatures can stratify patients with heterogeneous diseases, such as systemic lupus erythematosus (SLE), yet understanding the contributions of ancestral background to this heterogeneity is not well understood. We hypothesized that ancestry would significantly influence gene expression signatures and measured 34 gene modules in 1566 SLE patients of African ancestry (AA), European ancestry (EA), or Native American ancestry (NAA). Healthy subject ancestry-specific gene expression provided the transcriptomic background upon which the SLE patient signatures were built. Although standard therapy affected every gene signature and significantly increased myeloid cell signatures, logistic regression analysis determined that ancestral background significantly changed 23 of 34 gene signatures. Additionally, the strongest association to gene expression changes was found with autoantibodies, and this also had etiology in ancestry: the AA predisposition to have both RNP and dsDNA autoantibodies compared with EA predisposition to have only anti-dsDNA. A machine learning approach was used to determine a gene signature characteristic to distinguish AA SLE and was most influenced by genes characteristic of the perturbed B cell axis in AA SLE patients.


Asunto(s)
Biomarcadores/análisis , Negro o Afroamericano/genética , Predisposición Genética a la Enfermedad , Lupus Eritematoso Sistémico/patología , Polimorfismo de Nucleótido Simple , Nivel de Atención , Población Blanca/genética , Adulto , Autoanticuerpos/sangre , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Genotipo , Humanos , Lupus Eritematoso Sistémico/clasificación , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/terapia , Masculino , Persona de Mediana Edad , Pronóstico
10.
J Autoimmun ; 110: 102359, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31806421

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic, systemic autoimmune disease that causes damage to multiple organ systems. Despite decades of research and available murine models that capture some aspects of the human disease, new treatments for SLE lag behind other autoimmune diseases such as Rheumatoid Arthritis and Crohn's disease. Big data genomic assays have transformed our understanding of SLE by providing important insights into the molecular heterogeneity of this multigenic disease. Gene wide association studies have demonstrated more than 100 risk loci, supporting a model of multiple genetic hits increasing SLE risk in a non-linear fashion, and providing evidence of ancestral diversity in susceptibility loci. Epigenetic studies to determine the role of methylation, acetylation and non-coding RNAs have provided new understanding of the modulation of gene expression in SLE patients and identified new drug targets and biomarkers for SLE. Gene expression profiling has led to a greater understanding of the role of myeloid cells in the pathogenesis of SLE, confirmed roles for T and B cells in SLE, promoted clinical trials based on the prominent interferon signature found in SLE patients, and identified candidate biomarkers and cellular signatures to further drug development and drug repurposing. Gene expression studies are advancing our understanding of the underlying molecular heterogeneity in SLE and providing hope that patient stratification will expedite new therapies based on personal molecular signatures. Although big data analyses present unique interpretation challenges, both computationally and biologically, advances in machine learning applications may facilitate the ability to predict changes in SLE disease activity and optimize therapeutic strategies.


Asunto(s)
Susceptibilidad a Enfermedades , Lupus Eritematoso Sistémico/etiología , Lupus Eritematoso Sistémico/metabolismo , Alelos , Animales , Macrodatos , Biomarcadores , Minería de Datos , Susceptibilidad a Enfermedades/inmunología , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Lupus Eritematoso Sistémico/diagnóstico , Lupus Eritematoso Sistémico/terapia , Aprendizaje Automático , Medicina de Precisión/métodos
11.
Front Immunol ; 10: 2512, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31708928

RESUMEN

Autoantibody production by plasma cells (PCs) plays a pivotal role in the pathogenesis of systemic lupus erythematosus (SLE). The molecular pathways by which B cells become pathogenic PC secreting autoantibodies in SLE are incompletely characterized. Histone deactylase 6 (HDAC6) is a unique cytoplasmic HDAC that modifies the interaction of a number of tubulin- associated proteins; inhibition of HDAC6 has been shown to be beneficial in murine models of SLE, but the downstream pathways accounting for the therapeutic benefit have not been clearly delineated. In the current study, we sought to determine whether selective HDAC6 inhibition would abrogate abnormal B cell activation in SLE. We treated NZB/W lupus mice with the selective HDAC6 inhibitor, ACY-738, for 4 weeks beginning at 20 weeks-of age. After only 4 weeks of treatment, manifestation of lupus nephritis (LN) were greatly reduced in these animals. We then used RNAseq to determine the genomic signatures of splenocytes from treated and untreated mice and applied computational cellular and pathway analysis to reveal multiple signaling events associated with B cell activation and differentiation in SLE that were modulated by HDAC6 inhibition. PC development was abrogated and germinal center (GC) formation was greatly reduced. When the HDAC6 inhibitor-treated lupus mouse gene signatures were compared to human lupus patient gene signatures, the results showed numerous immune, and inflammatory pathways increased in active human lupus were significantly decreased in the HDAC6 inhibitor treated animals. Pathway analysis suggested alterations in cellular metabolism might contribute to the normalization of lupus mouse spleen genomic signatures, and this was confirmed by direct measurement of the impact of the HDAC6 inhibitor on metabolic activities of murine spleen cells. Taken together, these studies show HDAC6 inhibition decreases B cell activation signaling pathways and reduces PC differentiation in SLE and suggest that a critical event might be modulation of cellular metabolism.


Asunto(s)
Linfocitos B/efectos de los fármacos , Centro Germinal/inmunología , Histona Desacetilasa 6/antagonistas & inhibidores , Ácidos Hidroxámicos/farmacología , Lupus Eritematoso Sistémico/tratamiento farmacológico , Activación de Linfocitos/efectos de los fármacos , Pirimidinas/farmacología , Animales , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Femenino , Lupus Eritematoso Sistémico/inmunología , Ratones , Transducción de Señal/efectos de los fármacos
12.
Front Immunol ; 10: 2136, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31616406

RESUMEN

Autoimmune diseases (AID) such as systemic lupus erythematosus (SLE), primary Sjögren's syndrome (pSS), and rheumatoid arthritis (RA) are chronic inflammatory diseases in which abnormalities of B cell function play a central role. Although it is widely accepted that autoimmune B cells are hyperactive in vivo, a full understanding of their functional status in AID has not been delineated. Here, we present a detailed analysis of the functional capabilities of AID B cells and dissect the mechanisms underlying altered B cell function. Upon BCR activation, decreased spleen tyrosine kinase (Syk) and Bruton's tyrosine kinase (Btk) phosphorylation was noted in AID memory B cells combined with constitutive co-localization of CD22 and protein tyrosine phosphatase (PTP) non-receptor type 6 (SHP-1) along with hyporesponsiveness to TLR9 signaling, a Syk-dependent response. Similar BCR hyporesponsiveness was also noted specifically in SLE CD27- B cells together with increased PTP activities and increased transcripts for PTPN2, PTPN11, PTPN22, PTPRC, and PTPRO in SLE B cells. Additional studies revealed that repetitive BCR stimulation of normal B cells can induce BCR hyporesponsiveness and that tissue-resident memory B cells from AID patients also exhibited decreased responsiveness immediately ex vivo, suggesting that the hyporesponsive status can be acquired by repeated exposure to autoantigen(s) in vivo. Functional studies to overcome B cell hyporesponsiveness revealed that CD40 co-stimulation increased BCR signaling, induced proliferation, and downregulated PTP expression (PTPN2, PTPN22, and receptor-type PTPs). The data support the conclusion that hyporesponsiveness of AID and especially SLE B cells results from chronic in vivo stimulation through the BCR without T cell help mediated by CD40-CD154 interaction and is manifested by decreased phosphorylation of BCR-related proximal signaling molecules and increased PTPs. The hyporesponsiveness of AID B cells is similar to a form of functional anergy.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos B/inmunología , Agammaglobulinemia Tirosina Quinasa/inmunología , Humanos , Proteínas Tirosina Fosfatasas/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Quinasa Syk/inmunología
13.
Sci Rep ; 9(1): 9617, 2019 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-31270349

RESUMEN

The integration of gene expression data to predict systemic lupus erythematosus (SLE) disease activity is a significant challenge because of the high degree of heterogeneity among patients and study cohorts, especially those collected on different microarray platforms. Here we deployed machine learning approaches to integrate gene expression data from three SLE data sets and used it to classify patients as having active or inactive disease as characterized by standard clinical composite outcome measures. Both raw whole blood gene expression data and informative gene modules generated by Weighted Gene Co-expression Network Analysis from purified leukocyte populations were employed with various classification algorithms. Classifiers were evaluated by 10-fold cross-validation across three combined data sets or by training and testing in independent data sets, the latter of which amplified the effects of technical variation. A random forest classifier achieved a peak classification accuracy of 83 percent under 10-fold cross-validation, but its performance could be severely affected by technical variation among data sets. The use of gene modules rather than raw gene expression was more robust, achieving classification accuracies of approximately 70 percent regardless of how the training and testing sets were formed. Fine-tuning the algorithms and parameter sets may generate sufficient accuracy to be informative as a standalone estimate of disease activity.


Asunto(s)
Biología Computacional , Perfilación de la Expresión Génica , Expresión Génica , Lupus Eritematoso Sistémico/genética , Aprendizaje Automático , Biología Computacional/métodos , Progresión de la Enfermedad , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Lupus Eritematoso Sistémico/diagnóstico , Anotación de Secuencia Molecular , Curva ROC , Transcriptoma
14.
Commun Biol ; 2: 140, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31044165

RESUMEN

A role for interferon (IFN) in systemic lupus erythematosus (SLE) pathogenesis is inferred from the prominent IFN gene signature (IGS), but the major IFN species and its relationship to disease activity are unknown. A bioinformatic approach employing individual IFN species gene signatures to interrogate SLE microarray datasets demonstrates a putative role for numerous IFN species, with prominent expression of IFNB1 and IFNW signatures. In contrast with other SLE-affected organs, the IGS is less prominent in lupus nephritis. SLE patients with active and inactive disease have readily detectable IGS and the IGS changes synchronously with a monocyte signature but not disease activity, and is significantly related to monocyte transcripts. Monocyte over-expression of three times as many IGS transcripts as T and B cells and IGS retention in monocytes, but not T and B cells from inactive SLE patients contribute to the lack of correlation between the IGS and SLE disease activity.


Asunto(s)
Interferones/genética , Lupus Eritematoso Sistémico/genética , Transcriptoma , Conjuntos de Datos como Asunto , Humanos , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Riñón/metabolismo , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/metabolismo , Nefritis Lúpica/genética , Nefritis Lúpica/metabolismo , Monocitos/metabolismo , Especificidad de Órganos , Piel/metabolismo , Membrana Sinovial/metabolismo , Análisis de Matrices Tisulares , Transcriptoma/efectos de los fármacos
15.
J Immunol ; 202(11): 3309-3317, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31019061

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the presence of low-density granulocytes (LDGs) with a heightened capacity for spontaneous NETosis, but the contribution of LDGs to SLE pathogenesis remains unclear. To characterize LDGs in human SLE, gene expression profiles derived from isolated LDGs were characterized by weighted gene coexpression network analysis, and a 92-gene module was identified. The LDG gene signature was enriched in genes related to neutrophil degranulation and cell cycle regulation. This signature was assessed in gene expression datasets from two large-scale SLE clinical trials to study associations between LDG enrichment, SLE manifestations, and treatment regimens. LDG enrichment in the blood was associated with corticosteroid treatment as well as anti-dsDNA, low serum complement, renal manifestations, and vasculitis, but the latter two of these associations were dependent on concomitant corticosteroid treatment. In addition, LDG enrichment was associated with enrichment of gene signatures induced by type I IFN and TNF irrespective of corticosteroid treatment. Notably, LDG enrichment was not found in numerous tissues affected by SLE. Comparison with relevant reference datasets indicated that LDG enrichment is likely reflective of increased granulopoiesis in the bone marrow and not peripheral neutrophil activation. The results have uncovered important determinants of the appearance of LDGs in SLE and have emphasized the likely role of LDGs in specific aspects of lupus pathogenesis.


Asunto(s)
Trampas Extracelulares/inmunología , Granulocitos/fisiología , Lupus Eritematoso Sistémico/genética , Corticoesteroides/uso terapéutico , Anticuerpos Antinucleares/sangre , Ciclo Celular/genética , Degranulación de la Célula/genética , Conjuntos de Datos como Asunto , Redes Reguladoras de Genes , Genómica , Hematopoyesis , Humanos , Interferón Tipo I/metabolismo , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Activación Neutrófila , Transcriptoma
16.
PLoS One ; 13(12): e0208132, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30562343

RESUMEN

Systemic lupus erythematosus (SLE) is characterized by abnormalities in B cell and T cell function, but the role of disturbances in the activation status of macrophages (Mϕ) has not been well described in human patients. To address this, gene expression profiles from isolated lymphoid and myeloid populations were analyzed to identify differentially expressed (DE) genes between healthy controls and patients with either inactive or active SLE. While hundreds of DE genes were identified in B and T cells of active SLE patients, there were no DE genes found in B or T cells from patients with inactive SLE compared to healthy controls. In contrast, large numbers of DE genes were found in myeloid cells (MC) from both active and inactive SLE patients. Among the DE genes were several known to play roles in Mϕ activation and polarization, including the M1 genes STAT1 and SOCS3 and the M2 genes STAT3, STAT6, and CD163. M1-associated genes were far more frequent in data sets from active versus inactive SLE patients. To characterize the relationship between Mϕ activation and disease activity in greater detail, weighted gene co-expression network analysis (WGCNA) was used to identify modules of genes associated with clinical activity in SLE patients. Among these were disease activity-correlated modules containing activation signatures of predominantly M1-associated genes. No disease activity-correlated modules were enriched in M2-associated genes. Pathway and upstream regulator analysis of DE genes from both active and inactive SLE MC were cross-referenced with high-scoring hits from the drug discovery Library of Integrated Network-based Cellular Signatures (LINCS) to identify new strategies to treat both stages of SLE. A machine learning approach employing MC gene modules and a generalized linear model was able to predict the disease activity status in unrelated gene expression data sets. In summary, altered MC gene expression is characteristic of both active and inactive SLE. However, disease activity is associated with an alteration in the activation of MC, with a bias toward the M1 proinflammatory phenotype. These data suggest that while hyperactivity of B cells and T cells is associated with active SLE, MC potentially direct flare-ups and remission by altering their activation status toward the M1 state.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Lupus Eritematoso Sistémico/inmunología , Activación de Macrófagos/genética , Macrófagos/inmunología , Biología Computacional , Conjuntos de Datos como Asunto , Perfilación de la Expresión Génica , Redes Reguladoras de Genes/inmunología , Humanos , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/genética , Aprendizaje Automático , Macrófagos/metabolismo , Brote de los Síntomas , Transcriptoma/inmunología
18.
J Immunol ; 172(5): 3297-304, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14978138

RESUMEN

We report on the role of specific CD8(+) T cells in the pathogenesis of a highly lethal human viral disease, hantavirus pulmonary syndrome (HPS). HPS is a zoonotic disease caused by transmission of Sin Nombre virus (SNV) from chronically infected deer mice. In humans, this fulminant infection is characterized by lung capillary leakage, respiratory failure, and cardiogenic shock. Individuals with HLA-B*3501 have an increased risk of developing severe HPS, suggesting that CD8(+) T cell responses to SNV contribute to pathogenesis. We identified three CD8(+) T cell epitopes in SNV presented by HLA-B*3501 and quantitated circulating SNV-specific CD8(+) T cells in 11 acute HPS patients using HLA/peptide tetramers. We found significantly higher frequencies of SNV-specific T cells in patients with severe HPS requiring mechanical ventilation (up to 44.2% of CD8(+) T cells) than in moderately ill HPS patients hospitalized but not requiring mechanical ventilation (up to 9.8% of CD8(+) T cells). These results imply that virus-specific CD8(+) T cells contribute to HPS disease outcome. Intense CD8(+) T cell responses to SNV may be induced by the encounter of the unnatural human host to this zoonotic virus without coevolution. This may also be the immunopathologic basis of other life-threatening human virus infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Síndrome Pulmonar por Hantavirus/inmunología , Fiebres Hemorrágicas Virales/inmunología , Zoonosis/virología , Secuencia de Aminoácidos , Animales , Linfocitos T CD8-positivos/virología , Epítopos de Linfocito T/análisis , Síndrome Pulmonar por Hantavirus/virología , Fiebres Hemorrágicas Virales/virología , Humanos , Cinética , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Recuento de Linfocitos , Datos de Secuencia Molecular , Índice de Severidad de la Enfermedad , Virus Sin Nombre/inmunología
19.
J Immunol ; 168(8): 4184-91, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11937579

RESUMEN

High frequencies of EBV-specific CD8(+) T cells have been detected during acute EBV infection, yet persistent infection inevitably results. To address this issue, we characterized the phenotype and function of epitope-specific CD8(+) T cell populations from presentation with acute through latent infection. Considerable phenotypic and functional heterogeneity within, as well as between, two different epitope-specific populations was observed over time following acute infection. B7 EBV-encoded nuclear Ag (EBNA)-3A-specific CD8(+) T cells expressed only CD45RO from acute through latent EBV infection. A2 BMLF-1-specific CD8(+) T cells expressed CD45RO during acute infection and either CD45RA or CD45RO during latent EBV infection. This difference in CD45 isoform expression between the two epitope-specific populations did not translate into differences in perforin content, the ability to produce IFN-gamma, or the ability to proliferate in response to Ag in vitro. In individuals with latent EBV infection, the frequencies of A2 BMLF-1- or B7 EBNA-3A-specific CD8(+) T cells that expressed CD45RA, CD45RO, CD62 ligand, CCR7, and perforin were stable over time. However, the expression of CD62 ligand and CCR7 was significantly higher among EBNA-3A-specific CD8(+) T cells than among BMLF-1-specific CD8(+) T cells. Further work is necessary to understand how phenotypic and functional differences between EBV epitope-specific CD8(+) T cells are related to the biology of the virus and to the equilibrium between the virus and the host during persistent infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/biosíntesis , Epítopos de Linfocito T/inmunología , Herpesvirus Humano 4/inmunología , Inmunofenotipificación , Subgrupos de Linfocitos T/inmunología , Adolescente , Adulto , Antígeno B7-1/metabolismo , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Línea Celular Transformada , Membrana Celular/enzimología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Antígeno HLA-A2/metabolismo , Humanos , Memoria Inmunológica , Interferón gamma/biosíntesis , Isoenzimas/biosíntesis , Antígenos Comunes de Leucocito/biosíntesis , Activación de Linfocitos , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/metabolismo , Oligopéptidos/metabolismo , Perforina , Proteínas Citotóxicas Formadoras de Poros , Unión Proteica/inmunología , Receptores Mensajeros de Linfocitos/biosíntesis , Subgrupos de Linfocitos T/enzimología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...