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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38841857

RESUMEN

BACKGROUND: Pulmonary hypertension (PH) is a common complication of systemic sclerosis (SSc) and a leading cause of mortality among patients with this disease. PH can also occur as an idiopathic condition (idiopathic pulmonary arterial hypertension). Investigation of transcriptomic alterations in vascular populations is critical to elucidating cellular mechanisms underlying pathobiology of SSc-associated and idiopathic PH. METHODS: We analyzed single-cell RNA sequencing profiles of endothelial and perivascular mesenchymal populations from explanted lung tissue of patients with SSc-associated PH (n=16), idiopathic pulmonary arterial hypertension (n=3), and healthy controls (n=15). Findings were validated by immunofluorescence staining of explanted human lung tissue. RESULTS: Three disease-associated endothelial populations emerged. Two angiogenic endothelial cell (EC) subtypes markedly expanded in SSc-associated PH lungs: tip ECs expressing canonical tip markers PGF and APLN and phalanx ECs expressing genes associated with vascular development, endothelial barrier integrity, and Notch signaling. Gene regulatory network analysis suggested enrichment of Smad1 and PPAR-γ (peroxisome proliferator-activated receptor-γ) regulon activities in these 2 populations, respectively. Mapping of potential ligand-receptor interactions highlighted Notch, apelin-APJ, and angiopoietin-Tie signaling pathways between angiogenic ECs and perivascular cells. Transitional cells, expressing both endothelial and pericyte/smooth muscle cell markers, provided evidence for the presence of endothelial-to-mesenchymal transition. Transcriptional programs associated with arterial endothelial dysfunction implicated VEGF-A (vascular endothelial growth factor-A), TGF-ß1, angiotensin, and TNFSF12/TWEAK in the injury/remodeling phenotype of PH arterial ECs. CONCLUSIONS: These data provide high-resolution insights into the complexity and plasticity of the pulmonary endothelium in SSc-associated PH and idiopathic pulmonary arterial hypertension and provide direct molecular insights into soluble mediators and transcription factors driving PH vasculopathy.

2.
Am J Physiol Lung Cell Mol Physiol ; 323(4): L484-L494, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35997276

RESUMEN

Accumulation of excessive extracellular matrix (ECM) components from lung fibroblasts is a feature of systemic sclerosis-associated interstitial lung disease (SSc-ILD), and there is increasing evidence that innate immune signaling pathways contribute to these processes. Toll-like receptors (TLRs) are innate immune sensors activated by danger signals derived from pathogens or host molecular patterns. Several damage-associated molecular pattern (DAMP) molecules are elevated in SSc-ILD plasma, including ligands that activate TLR9, an innate immune sensor recently implicated in driving profibrotic responses in fibroblasts. Fibronectin and the isoform fibronectin-extra domain A (FN-EDA) are prominent in pathological extracellular matrix accumulation, but mechanisms promoting FN-EDA accumulation are only partially understood. Here, we show that TLR9 activation increases FN-EDA accumulation in MRC5 and SSc-ILD fibroblasts, but that this effect is independent of changes in FN-EDA gene transcription. Rather, we describe a novel mechanism where TLR9 activation inhibits FN-EDA turnover via reduced FN-EDA ubiquitination. TLR9 ligand ODN2006 reduces ubiquitinated FN-EDA destined for lysosomal degradation, an effect abrogated with TLR9 knockdown or inhibition. Taken together, these results provide rationale for disrupting the TLR9 signaling axis or FN-EDA degradation pathways to reduce FN-EDA accumulation in SSc-ILD fibroblasts. More broadly, enhancing intracellular degradation of ECM components through TLR9 inhibition or enhanced ECM turnover could be a novel strategy to attenuate pathogenic ECM accumulation in SSc-ILD.


Asunto(s)
Fibronectinas , Enfermedades Pulmonares Intersticiales , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Humanos , Ligandos , Enfermedades Pulmonares Intersticiales/metabolismo , Isoformas de Proteínas/metabolismo , Receptor Toll-Like 9/genética , Ubiquitinación
3.
J Biol Chem ; 295(13): 4171-4180, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32071084

RESUMEN

Systemic scleroderma (SSc) is an autoimmune disease that affects over 2.5 million people globally. SSc results in dysfunctional connective tissues with excessive profibrotic signaling, affecting skin, cardiovascular, and particularly lung tissue. Over three-quarters of individuals with SSc develop pulmonary fibrosis within 5 years, the main cause of SSc mortality. No approved medicines to manage lung SSc currently exist. Recent research suggests that profibrotic signaling by transforming growth factor ß (TGF-ß) is directly tied to SSc. Previous studies have also shown that ubiquitin E3 ligases potently control TGF-ß signaling through targeted degradation of key regulatory proteins; however, the roles of these ligases in SSc-TGF-ß signaling remain unclear. Here we utilized primary SSc patient lung cells for high-throughput screening of TGF-ß signaling via high-content imaging of nuclear translocation of the profibrotic transcription factor SMAD family member 2/3 (SMAD2/3). We screened an RNAi library targeting ubiquitin E3 ligases and observed that knockdown of the E3 ligase Kelch-like protein 42 (KLHL42) impairs TGF-ß-dependent profibrotic signaling. KLHL42 knockdown reduced fibrotic tissue production and decreased TGF-ß-mediated SMAD activation. Using unbiased ubiquitin proteomics, we identified phosphatase 2 regulatory subunit B'ϵ (PPP2R5ϵ) as a KLHL42 substrate. Mechanistic experiments validated ubiquitin-mediated control of PPP2R5ϵ stability through KLHL42. PPP2R5ϵ knockdown exacerbated TGF-ß-mediated profibrotic signaling, indicating a role of PPP2R5ϵ in SSc. Our findings indicate that the KLHL42-PPP2R5ϵ axis controls profibrotic signaling in SSc lung fibroblasts. We propose that future studies could investigate whether chemical inhibition of KLHL42 may ameliorate profibrotic signaling in SSc.


Asunto(s)
Proteína Fosfatasa 2/genética , Esclerodermia Sistémica/genética , Proteína Smad2/genética , Factor de Crecimiento Transformador beta/genética , Ubiquitina-Proteína Ligasas/genética , Fibroblastos/metabolismo , Fibrosis/genética , Fibrosis/patología , Humanos , Pulmón/citología , Pulmón/metabolismo , Proteolisis , Proteómica , Esclerodermia Sistémica/patología , Transducción de Señal/genética
4.
Eur Respir J ; 54(2)2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31221805

RESUMEN

A comprehensive understanding of the changes in gene expression in cell types involved in idiopathic pulmonary fibrosis (IPF) will shed light on the mechanisms underlying the loss of alveolar epithelial cells and development of honeycomb cysts and fibroblastic foci. We sought to understand changes in IPF lung cell transcriptomes and gain insight into innate immune aspects of pathogenesis.We investigated IPF pathogenesis using single-cell RNA-sequencing of fresh lung explants, comparing human IPF fibrotic lower lobes reflecting late disease, upper lobes reflecting early disease and normal lungs.IPF lower lobes showed increased fibroblasts, and basal, ciliated, goblet and club cells, but decreased alveolar epithelial cells, and marked alterations in inflammatory cells. We found three discrete macrophage subpopulations in normal and fibrotic lungs, one expressing monocyte markers, one highly expressing FABP4 and INHBA (FABP4hi), and one highly expressing SPP1 and MERTK (SPP1hi). SPP1hi macrophages in fibrotic lower lobes showed highly upregulated SPP1 and MERTK expression. Low-level local proliferation of SPP1hi macrophages in normal lungs was strikingly increased in IPF lungs.Co-localisation and causal modelling supported the role for these highly proliferative SPP1hi macrophages in activation of IPF myofibroblasts in lung fibrosis. These data suggest that SPP1hi macrophages contribute importantly to lung fibrosis in IPF, and that therapeutic strategies targeting MERTK and macrophage proliferation may show promise for treatment of this disease.


Asunto(s)
Fibrosis Pulmonar Idiopática/metabolismo , Macrófagos/metabolismo , Osteopontina/metabolismo , Tirosina Quinasa c-Mer/metabolismo , Proliferación Celular , Células Epiteliales/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/genética , Sistema Inmunológico , Inmunidad Innata , Subunidades beta de Inhibinas/metabolismo , Pulmón/metabolismo , Miofibroblastos/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Procesos Estocásticos
5.
Ann Rheum Dis ; 78(10): 1379-1387, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31405848

RESUMEN

OBJECTIVES: Myofibroblasts are key effector cells in the extracellular matrix remodelling of systemic sclerosis-associated interstitial lung disease (SSc-ILD); however, the diversity of fibroblast populations present in the healthy and SSc-ILD lung is unknown and has prevented the specific study of the myofibroblast transcriptome. We sought to identify and define the transcriptomes of myofibroblasts and other mesenchymal cell populations in human healthy and SSc-ILD lungs to understand how alterations in fibroblast phenotypes lead to SSc-ILD fibrosis. METHODS: We performed droplet-based, single-cell RNA-sequencing with integrated canonical correlation analysis of 13 explanted lung tissue specimens (56 196 cells) from four healthy control and four patients with SSc-ILD, with findings confirmed by cellular indexing of transcriptomes and epitopes by sequencing in additional samples. RESULTS: Examination of gene expression in mesenchymal cells identified two major, SPINT2hi and MFAP5hi, and one minor, WIF1hi, fibroblast populations in the healthy control lung. Combined analysis of control and SSc-ILD mesenchymal cells identified SPINT2hi, MFAP5hi, few WIF1hi fibroblasts and a new large myofibroblast population with evidence of actively proliferating myofibroblasts. We compared differential gene expression between all SSc-ILD and control mesenchymal cell populations, as well as among the fibroblast subpopulations, showing that myofibroblasts undergo the greatest phenotypic changes in SSc-ILD and strongly upregulate expression of collagens and other profibrotic genes. CONCLUSIONS: Our results demonstrate previously unrecognised fibroblast heterogeneity in SSc-ILD and healthy lungs, and define multimodal transcriptome-phenotypes associated with these populations. Our data indicate that myofibroblast differentiation and proliferation are key pathological mechanisms driving fibrosis in SSc-ILD.


Asunto(s)
Fibroblastos/metabolismo , Heterogeneidad Genética , Enfermedades Pulmonares Intersticiales/genética , Miofibroblastos/metabolismo , Esclerodermia Sistémica/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Estudios de Casos y Controles , Proliferación Celular/genética , Colágeno/genética , Proteínas Contráctiles/metabolismo , Epítopos , Femenino , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Pulmón/citología , Masculino , Glicoproteínas de Membrana/metabolismo , Mesodermo/citología , Persona de Mediana Edad , Fenotipo , Fibrosis Pulmonar/genética , Esclerodermia Sistémica/complicaciones , Análisis de la Célula Individual , Transcriptoma
6.
J Immunol ; 192(12): 5471-5475, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24829419

RESUMEN

We reported previously that c-kit ligation by membrane-bound stem cell factor (mSCF) boosts IL-6 production in dendritic cells (DCs) and a Th17-immune response. However, Th17 establishment also requires heterodimeric IL-23, but the mechanisms that regulate IL-23 gene expression in DCs are not fully understood. We show that IL-23p19 gene expression in lung DCs is dependent on mSCF, which is regulated by the metalloproteinase MMP-9. Th1-inducing conditions enhanced MMP-9 activity, causing cleavage of mSCF, whereas the opposite was true for Th17-promoting conditions. In MMP-9(-/-) mice, a Th1-inducing condition could maintain mSCF and enhance IL-23p19 in DCs, promoting IL-17-producing CD4(+) T cells in the lung. Conversely, mSCF cleavage from bone marrow DCs in vitro by rMMP-9 led to reduced IL-23p19 expression under Th17-inducing conditions, with dampening of intracellular AKT phosphorylation. Collectively, these results show that the c-kit/mSCF/MMP-9 axis regulates IL-23 gene expression in DCs to control IL-17 production in the lung.


Asunto(s)
Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Interleucina-17/inmunología , Subunidad p19 de la Interleucina-23/inmunología , Pulmón/inmunología , Metaloproteinasa 9 de la Matriz/inmunología , Factor de Células Madre/inmunología , Animales , Membrana Celular/genética , Membrana Celular/inmunología , Membrana Celular/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica/genética , Interleucina-17/genética , Interleucina-17/metabolismo , Subunidad p19 de la Interleucina-23/biosíntesis , Subunidad p19 de la Interleucina-23/genética , Pulmón/citología , Pulmón/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Fosforilación/genética , Fosforilación/inmunología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Células Madre/genética , Factor de Células Madre/metabolismo , Células TH1/citología , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/citología , Células Th17/inmunología , Células Th17/metabolismo
7.
Arthritis Rheumatol ; 74(2): 329-341, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34042322

RESUMEN

OBJECTIVE: In this study, we sought a comprehensive understanding of myeloid cell types driving fibrosis in diffuse cutaneous systemic sclerosis (dcSSc) skin. METHODS: We analyzed the transcriptomes of 2,465 myeloid cells from skin biopsy specimens from 12 dcSSc patients and 10 healthy control subjects using single-cell RNA sequencing. Monocyte-derived dendritic cells (mo-DCs) were assessed using immunohistochemical staining and immunofluorescence analyses targeting ficolin-1 (FCN-1). RESULTS: A t-distributed stochastic neighbor embedding analysis of single-cell transcriptome data revealed 12 myeloid cell clusters, 9 of which paralleled previously described healthy control macrophage/DC clusters, and 3 of which were dcSSc-specific myeloid cell clusters. One SSc-associated macrophage cluster, highly expressing Fcγ receptor IIIA, was suggested on pseudotime analysis to be derived from normal CCR1+ and MARCO+ macrophages. A second SSc-associated myeloid population highly expressed monocyte markers FCN-1, epiregulin, S100A8, and S100A9, but was closely related to type 2 conventional DCs on pseudotime analysis and identified as mo-DCs. Mo-DCs were associated with more severe skin disease. Proliferating macrophages and plasmacytoid DCs were detected almost exclusively in dcSSc skin, the latter clustering with B cells and apparently derived from lymphoid progenitors. CONCLUSION: Transcriptional signatures in these and other myeloid populations indicate innate immune system activation, possibly through Toll-like receptors and highly up-regulated chemokines. However, the appearance and activation of myeloid cells varies between patients, indicating potential differences in the underlying pathogenesis and/or temporal disease activity in dcSSc.


Asunto(s)
Células Dendríticas/inmunología , Lectinas/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Receptores de IgG/inmunología , Esclerodermia Difusa/inmunología , Humanos , Lectinas/biosíntesis , Macrófagos/metabolismo , Monocitos/metabolismo , Receptores de IgG/biosíntesis , Índice de Severidad de la Enfermedad , Ficolinas
8.
Arthritis Rheumatol ; 74(12): 2003-2014, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35849803

RESUMEN

OBJECTIVE: Systemic sclerosis-associated interstitial lung disease (SSc-ILD) is the leading cause of death in patients with SSc with unclear pathogenesis and limited treatment options. Evidence strongly supports an important role for profibrotic secreted phosphoprotein 1 (SPP1)-expressing macrophages in SSc-ILD. This study was undertaken to define the transcriptome and chromatin structural changes of SPP1 SSc-ILD macrophages in order to better understand their role in promoting fibrosis and to identify transcription factors associated with open chromatin driving their altered phenotype. METHODS: We performed single-cell RNA sequencing (scRNA-Seq) on 11 explanted SSc-ILD and healthy control lung samples, as well as single-cell assay for transposase-accessible chromatin sequencing on 5 lung samples to define altered chromatin accessibility of SPP1 macrophages. We predicted transcription factors regulating SPP1 macrophages using single-cell regulatory network inference and clustering (SCENIC) and determined transcription factor binding sites associated with global alterations in SPP1 chromatin accessibility using Signac/Seurat. RESULTS: We identified distinct macrophage subpopulations using scRNA-Seq analysis in healthy and SSc-ILD lungs and assessed gene expression changes during the change of healthy control macrophages into SPP1 macrophages. Analysis of open chromatin validated SCENIC predictions, indicating that microphthalmia-associated transcription factor, transcription factor EB, activating transcription factor 6, sterol regulatory element binding transcription factor 1, basic helix-loop-helix family member E40, Kruppel-like factor 6, ETS variant transcription factor 5, and/or members of the activator protein 1 family of transcription factors regulate SPP1 macrophage differentiation. CONCLUSION: Our findings shed light on the underlying changes in chromatin structure and transcription factor regulation of profibrotic SPP1 macrophages in SSc-ILD. Similar alterations in SPP1 macrophages may underpin fibrosis in other organs involved in SSc and point to novel targets for the treatment of SSc-ILD, specifically targeting profibrotic macrophages.


Asunto(s)
Enfermedades Pulmonares Intersticiales , Esclerodermia Sistémica , Humanos , Enfermedades Pulmonares Intersticiales/genética , Enfermedades Pulmonares Intersticiales/patología , Epigénesis Genética , Esclerodermia Sistémica/complicaciones , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/patología , Macrófagos/metabolismo , Pulmón/patología , Fibrosis , Factores de Transcripción/genética , Cromatina
9.
J Scleroderma Relat Disord ; 6(3): 242-246, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-35005243

RESUMEN

INTRODUCTION: Given the poor treatment options for pulmonary arterial hypertension associated systemic sclerosis (SSc-PAH) patients, we sought to determine clinical safety and efficacy of Dimethylfumarate (DMF), an Nrf2 agonist, and the effects on biomarkers of oxidative stress on SSc-PAH in an exploratory interventional clinical trial. OBJECTIVES: The primary objectives were to assess the safety and efficacy of treatment with DMF in patients with SSc-PAH. METHODS: This was an investigator-initiated, double-blind, randomized, placebo-controlled trial conducted at two sites in the United States. The primary safety endpoint was the incidence of serious adverse events (SAEs) and all adverse events (AEs) in DMF compared to placebo-treated patients. The primary efficacy endpoint was the change in 6MWD from baseline to the end of treatment at Week 24 in DMF compared to placebo-treated patients. RESULTS: Six participants were randomized to either placebo (n = 2) or DMF (n = 4). Baseline demographics were similar in both groups. A total of 25 adverse events (AEs) occurred in 6 subjects, with 14 AEs (56.0%) having occurred in DMF-treated subjects. 3 occurrences were identified as nausea AEs, and two participants withdrew due to nausea. One participant in the placebo group was withdrawn after a hospitalization SAE due to worsening of heart failure and shortness of breath secondary to anemia. One participant in each group completed protocol. Subjects in the DMF-treated group showed a non-significant reduced decline in 6MWD (relative mean change of -7.07%) from baseline to Week 24 as compared to placebo-treated subjects (relative mean change of -14.97%). CONCLUSION: Patients treated for SSc-PAH with 2 and 3-drug regimens, as is now typical for these patients, tolerate DMF poorly. Our small samples size did not provide power to suggest efficacy. We suggest that Nrf2 is still a valid therapeutic target for future trials, using better tolerated Nrf2 agonists.

10.
J Dermatol Sci ; 97(1): 41-49, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31836271

RESUMEN

BACKGROUND: The heterogeneous functions of dermal myeloid cells in antigen presentation, and scavenging pathogens and cell debris places them centrally in cutaneous inflammation. Single cell transcriptomics can provide new understanding of the heterogeneity and function of yet incompletely understood human dermal myeloid cell subsets. OBJECTIVE: Investigate the transcriptome landscape of myeloid cells in healthy human skin. METHODS: Single cell RNA-sequencing was performed on skin biopsies from ten healthy donors and analyzed to identify myeloid cell populations. RESULTS: One LIN- HLA-DR+ cluster with expression of myeloid-specific genes was identified as a cluster of myeloid cells. Upon reanalysis of this cluster, we identified three macrophage subsets, marked by high expression of CCR1, MARCO or TREM2; and six dendritic cell subsets, marked by high expression of CLEC9A, CXorf21, MCOLN2, LAMP3, KIAA0101 and Langerin, representing respectively cDC1, two subsets of cDC2, a novel DC type, a cluster of proliferating DC, and a Langerhans cell subset. GO term analysis indicated specialized functions for the discrete rare populations of myeloid cells: TREM2 Mφ in lipid metabolism and LAMP3 DC as a mature cDC. Proliferating DCs appeared to represent cDC2 progenitors. CONCLUSION: The transcriptional landscape of myeloid cell populations in human skin indicates several, novel populations with specialized functions, as well as a rare proliferating DC population that likely accounts for local regeneration or expansion of dermal DCs. We provide robust gene expression markers for each of these populations that should permit better understandings of their roles in various homeostatic and pathologic immune processes in the skin.


Asunto(s)
Células Dendríticas/fisiología , Células Mieloides/metabolismo , Regeneración , Piel/citología , Transcriptoma/fisiología , Biopsia , Proliferación Celular/genética , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , RNA-Seq , Análisis de la Célula Individual , Adulto Joven
11.
J Invest Dermatol ; 138(4): 802-810, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29080679

RESUMEN

Fibroblasts produce matrix, regulate inflammation, mediate reparative processes, and serve as pluripotent mesenchymal cells. Analyzing digested normal human skin by single-cell RNA sequencing, we explored different fibroblast populations. T-distributed stochastic neighbor embedding and clustering of single-cell RNA sequencing data from six biopsy samples showed two major fibroblast populations, defined by distinct genes, including SFRP2 and FMO1, expressed exclusively by these two major fibroblast populations. Further subpopulations were defined within each of the SFRP2 and FMO1 populations and five minor fibroblast populations, each expressing discrete genes: CRABP1, COL11A1, FMO2, PRG4, or C2ORF40. Immunofluorescent staining confirmed that SFRP2 and FMO1 define cell types of dramatically different morphology. SFRP2+ fibroblasts were small, elongated, and distributed between collagen bundles. FMO1+ fibroblasts were larger and distributed in both interstitial and perivascular locations. Differential gene expression by SFRP2+, FMO1+, and COL11A1+ fibroblasts suggests roles in matrix deposition, inflammatory cell retention, and connective tissue cell differentiation, respectively.


Asunto(s)
Dipeptidil Peptidasa 4/genética , Regulación de la Expresión Génica , Proteínas de la Membrana/genética , Proteínas de Microfilamentos/genética , Oxigenasas/genética , Piel/citología , Animales , Antioxidantes , Diferenciación Celular , Células Cultivadas , Dipeptidil Peptidasa 4/biosíntesis , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Proteínas de la Membrana/biosíntesis , Ratones , Proteínas de Microfilamentos/biosíntesis , Oxigenasas/biosíntesis , ARN/genética , Piel/metabolismo
12.
Front Immunol ; 9: 2191, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30327649

RESUMEN

Objective: The mechanisms that lead to endothelial cell (EC) injury and propagate the vasculopathy in Systemic Sclerosis (SSc) are not well understood. Using single cell RNA sequencing (scRNA-seq), our goal was to identify EC markers and signature pathways associated with vascular injury in SSc skin. Methods: We implemented single cell sorting and subsequent RNA sequencing of cells isolated from SSc and healthy control skin. We used t-distributed stochastic neighbor embedding (t-SNE) to identify the various cell types. We performed pathway analysis using Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis (IPA). Finally, we independently verified distinct markers using immunohistochemistry on skin biopsies and qPCR in primary ECs from SSc and healthy skin. Results: By combining the t-SNE analysis with the expression of known EC markers, we positively identified ECs among the sorted cells. Subsequently, we examined the differential expression profile between the ECs from healthy and SSc skin. Using GSEA and IPA analysis, we demonstrated that the SSc endothelial cell expression profile is enriched in processes associated with extracellular matrix generation, negative regulation of angiogenesis and epithelial-to-mesenchymal transition. Two of the top differentially expressed genes, HSPG2 and APLNR, were independently verified using immunohistochemistry staining and real-time qPCR analysis. Conclusion: ScRNA-seq, differential gene expression and pathway analysis revealed that ECs from SSc patients show a discrete pattern of gene expression associated with vascular injury and activation, extracellular matrix generation and negative regulation of angiogenesis. HSPG2 and APLNR were identified as two of the top markers of EC injury in SSc.


Asunto(s)
Receptores de Apelina/genética , Receptores de Apelina/metabolismo , Células Endoteliales/metabolismo , Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Secuencia de Bases , Biomarcadores/metabolismo , Biopsia , Activación de Complemento , Transición Epitelial-Mesenquimal/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Expresión Génica , Humanos , Inmunohistoquímica , Metaloproteinasas de la Matriz/metabolismo , Neovascularización Patológica/metabolismo , Esclerodermia Sistémica/patología , Análisis de Secuencia de ARN , Lesiones del Sistema Vascular/patología
13.
JCI Insight ; 2(10)2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28515358

RESUMEN

Severe asthma (SA) is a significant problem both clinically and economically, given its poor response to corticosteroids (CS). We recently reported a complex type 1-dominated (IFN-γ-dominated) immune response in more than 50% of severe asthmatics despite high-dose CS treatment. Also, IFN-γ was found to be critical for increased airway hyperreactivity (AHR) in our model of SA. The transcription factor IRF5 expressed in M1 macrophages can induce a Th1/Th17 response in cocultured human T cells. Here we show markedly higher expression of IRF5 in bronchoalveolar lavage (BAL) cells of severe asthmatics as compared with that in cells from milder asthmatics or healthy controls. Using our SA mouse model, we demonstrate that lack of IRF5 in lymph node migratory DCs severely limits their ability to stimulate the generation of IFN-γ- and IL-17-producing CD4+ T cells and IRF5-/- mice subjected to the SA model displayed significantly lower IFN-γ and IL-17 responses, albeit showing a reciprocal increase in Th2 response. However, the absence of IRF5 rendered the mice responsive to CS with suppression of the heightened Th2 response. These data support the notion that IRF5 inhibition in combination with CS may be a viable approach to manage disease in a subset of severe asthmatics.

14.
Nat Commun ; 8: 13944, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28074841

RESUMEN

Bacterial pneumonia is a significant healthcare burden worldwide. Failure to resolve inflammation after infection precipitates lung injury and an increase in morbidity and mortality. Gram-negative bacteria are common in pneumonia and increased levels of the mito-damage-associated molecular pattern (DAMP) cardiolipin can be detected in the lungs. Here we show that mice infected with Klebsiella pneumoniae develop lung injury with accumulation of cardiolipin. Cardiolipin inhibits resolution of inflammation by suppressing production of anti-inflammatory IL-10 by lung CD11b+Ly6GintLy6CloF4/80+ cells. Cardiolipin induces PPARγ SUMOylation, which causes recruitment of a repressive NCOR/HDAC3 complex to the IL-10 promoter, but not the TNF promoter, thereby tipping the balance towards inflammation rather than resolution. Inhibition of HDAC activity by sodium butyrate enhances recruitment of acetylated histone 3 to the IL-10 promoter and increases the concentration of IL-10 in the lungs. These findings identify a mechanism of persistent inflammation during pneumonia and indicate the potential of HDAC inhibition as a therapy.


Asunto(s)
Cardiolipinas/fisiología , Inflamación/metabolismo , Interleucina-10/biosíntesis , Infecciones por Klebsiella/fisiopatología , Klebsiella pneumoniae/aislamiento & purificación , Neumonía Bacteriana/metabolismo , Animales , Cardiolipinas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Interleucina-10/genética , Interleucina-10/metabolismo , Infecciones por Klebsiella/microbiología , Lipopolisacáridos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Supresoras de Origen Mieloide/inmunología , Oxidación-Reducción , PPAR gamma/agonistas , PPAR gamma/metabolismo , Neumonía Bacteriana/tratamiento farmacológico , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/patología , Regiones Promotoras Genéticas , Células RAW 264.7 , Sumoilación , Factor de Necrosis Tumoral alfa/genética
15.
Cell Rep ; 15(8): 1700-14, 2016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27184852

RESUMEN

Inhalation of environmental antigens such as allergens does not always induce inflammation in the respiratory tract. While antigen-presenting cells (APCs), including dendritic cells and macrophages, take up inhaled antigens, the cell-intrinsic molecular mechanisms that prevent an inflammatory response during this process, such as activation of the transcription factor NF-κB, are not well understood. Here, we show that the nuclear receptor PPARγ plays a critical role in blocking NF-κB activation in response to inhaled antigens to preserve immune tolerance. Tolerance induction promoted mitochondrial respiration, generation of H2O2, and suppression of NF-κB activation in WT, but not PPARγ-deficient, APCs. Forced restoration of H2O2 in PPARγ-deficient cells suppressed IκBα degradation and NF-κB activation. Conversely, scavenging reactive oxygen species from mitochondria promoted IκBα degradation with loss of regulatory and promotion of inflammatory T cell responses in vivo. Thus, communication between PPARγ and the mitochondria maintains immune quiescence in the airways.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Peróxido de Hidrógeno/metabolismo , Pulmón/citología , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Animales , Antígeno CD11c/metabolismo , Proliferación Celular , Citocinas/genética , Células Dendríticas/metabolismo , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Tolerancia Inmunológica , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , PPAR gamma/deficiencia , PPAR gamma/metabolismo , Regiones Promotoras Genéticas/genética , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/citología
17.
J Clin Invest ; 125(8): 3037-50, 2015 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-26121748

RESUMEN

Severe asthma (SA) is a challenge to control, as patients are not responsive to high doses of systemic corticosteroids (CS). In contrast, mild-moderate asthma (MMA) is responsive to low doses of inhaled CS, indicating that Th2 cells, which are dominant in MMA, do not solely orchestrate SA development. Here, we analyzed broncholalveolar lavage cells isolated from MMA and SA patients and determined that IFN-γ (Th1) immune responses are exacerbated in the airways of individuals with SA, with reduced Th2 and IL-17 responses. We developed a protocol that recapitulates the complex immune response of human SA, including the poor response to CS, in a murine model. Compared with WT animals, Ifng-/- mice subjected to this SA model failed to mount airway hyperresponsiveness (AHR) without appreciable effect on airway inflammation. Conversely, AHR was not reduced in Il17ra-/- mice, although airway inflammation was lower. Computer-assisted pathway analysis tools linked IFN-γ to secretory leukocyte protease inhibitor (SLPI), which is expressed by airway epithelial cells, and IFN-γ inversely correlated with SLPI expression in SA patients and the mouse model. In mice subjected to our SA model, forced SLPI expression decreased AHR in the absence of CS, and it was further reduced when SLPI was combined with CS. Our study identifies a distinct immune response in SA characterized by a dysregulated IFN-γ/SLPI axis that affects lung function.


Asunto(s)
Asma/inmunología , Regulación de la Expresión Génica/inmunología , Interferón gamma/inmunología , Inhibidor Secretorio de Peptidasas Leucocitarias/inmunología , Células Th2/inmunología , Adolescente , Adulto , Animales , Asma/genética , Asma/patología , Lavado Broncoalveolar , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Humanos , Interferón gamma/genética , Interleucina-17/genética , Interleucina-17/inmunología , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Persona de Mediana Edad , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/inmunología , Inhibidor Secretorio de Peptidasas Leucocitarias/genética , Índice de Severidad de la Enfermedad , Células TH1/inmunología , Células TH1/patología , Células Th2/patología
18.
Hear Res ; 291(1-2): 34-40, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22743044

RESUMEN

The inferior colliculus (IC) of the big brown bat (Eptesicus fuscus) contains specialized neurons that respond exclusively to highly specific spectrotemporal patterns such as sinusoidally frequency modulated (SFM) signals or directional frequency modulated sweeps (FM). Other specialized cells with I-shaped frequency response areas (FRAs) are tuned to very narrow frequency bands (1-2 kHz) in an amplitude-tolerant manner. In contrast, non-specialized neurons respond to any stimulus with energy in their frequency response area. IC neurons in several mammalian species, including bats, demonstrate stimulus-specific adaptation (SSA), a reduction in response to a high-probability stimulus. To evaluate the relation between stimulus selectivity and SSA, we presented sounds using an oddball stimulus paradigm and recorded extracellular responses of IC neurons. SFM-selective cells (n = 10), FM-selective cells (n = 7), and cells with I-shaped FRAs (n = 13) did not show SSA under any of the conditions tested (NSSI = 0.009, 0.033, 0.020 respectively). However, non-specialized neurons (n = 52) exhibited various levels of SSA (NSSI = 0.163), with a subset of these cells displaying strong adaptation. These findings suggest that SSA is not a ubiquitous characteristic of all neurons in the bat IC, but is present only in a subset of non-specialized neurons.


Asunto(s)
Quirópteros/fisiología , Colículos Inferiores/fisiología , Estimulación Acústica , Adaptación Fisiológica , Animales , Conducta Animal , Quirópteros/anatomía & histología , Ecolocación/fisiología , Fenómenos Electrofisiológicos , Femenino , Colículos Inferiores/citología , Masculino , Neuronas/fisiología , Ratas , Especificidad de la Especie
19.
Nat Med ; 18(10): 1525-30, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22961107

RESUMEN

Immune tolerance is instituted early in life, during which time regulatory T (T(reg)) cells have an important role. Recurrent infections with respiratory syncytial virus (RSV) in early life increase the risk for asthma in adult life. Repeated infection of infant mice tolerized to ovalbumin (OVA) through their mother's milk with RSV induced allergic airway disease in response to OVA sensitization and challenge, including airway inflammation, hyper-reactivity and higher OVA-specific IgE, as compared to uninfected tolerized control mice. Virus infection induced GATA-3 expression and T helper type 2 (T(H)2) cytokine production in forkhead box P3 (FOXP3)(+) T(reg) cells and compromised the suppressive function of pulmonary T(reg) cells in a manner that was dependent on interleukin-4 receptor α (IL-4Rα) expression in the host. Thus, by promoting a T(H)2-type inflammatory response in the lung, RSV induced a T(H)2-like effector phenotype in T(reg) cells and attenuated tolerance to an unrelated antigen (allergen). Our findings highlight a mechanism by which viral infection targets a host-protective mechanism in early life and increases susceptibility to allergic disease.


Asunto(s)
Asma/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Linfocitos T Reguladores/inmunología , Animales , Asma/virología , Factores de Transcripción Forkhead/biosíntesis , Factor de Transcripción GATA3/biosíntesis , Tolerancia Inmunológica , Inmunoglobulina E/sangre , Subunidad alfa del Receptor de Interleucina-4/inmunología , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA