Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
2.
Nat Genet ; 56(4): 595-604, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38548990

RESUMEN

Common genetic variants confer substantial risk for chronic lung diseases, including pulmonary fibrosis. Defining the genetic control of gene expression in a cell-type-specific and context-dependent manner is critical for understanding the mechanisms through which genetic variation influences complex traits and disease pathobiology. To this end, we performed single-cell RNA sequencing of lung tissue from 66 individuals with pulmonary fibrosis and 48 unaffected donors. Using a pseudobulk approach, we mapped expression quantitative trait loci (eQTLs) across 38 cell types, observing both shared and cell-type-specific regulatory effects. Furthermore, we identified disease interaction eQTLs and demonstrated that this class of associations is more likely to be cell-type-specific and linked to cellular dysregulation in pulmonary fibrosis. Finally, we connected lung disease risk variants to their regulatory targets in disease-relevant cell types. These results indicate that cellular context determines the impact of genetic variation on gene expression and implicates context-specific eQTLs as key regulators of lung homeostasis and disease.


Asunto(s)
Fibrosis Pulmonar , Sitios de Carácter Cuantitativo , Humanos , Sitios de Carácter Cuantitativo/genética , Fibrosis Pulmonar/genética , Regulación de la Expresión Génica/genética , Pulmón , Herencia Multifactorial , Estudio de Asociación del Genoma Completo/métodos , Polimorfismo de Nucleótido Simple
3.
bioRxiv ; 2023 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-36993211

RESUMEN

Common genetic variants confer substantial risk for chronic lung diseases, including pulmonary fibrosis (PF). Defining the genetic control of gene expression in a cell-type-specific and context-dependent manner is critical for understanding the mechanisms through which genetic variation influences complex traits and disease pathobiology. To this end, we performed single-cell RNA-sequencing of lung tissue from 67 PF and 49 unaffected donors. Employing a pseudo-bulk approach, we mapped expression quantitative trait loci (eQTL) across 38 cell types, observing both shared and cell type-specific regulatory effects. Further, we identified disease-interaction eQTL and demonstrated that this class of associations is more likely to be cell-type specific and linked to cellular dysregulation in PF. Finally, we connected PF risk variants to their regulatory targets in disease-relevant cell types. These results indicate that cellular context determines the impact of genetic variation on gene expression, and implicates context-specific eQTL as key regulators of lung homeostasis and disease.

4.
Commun Biol ; 5(1): 1366, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36513703

RESUMEN

Cellular metabolism influences immune cell function, with mitochondrial fatty acid ß-oxidation and oxidative phosphorylation required for multiple immune cell phenotypes. Carnitine palmitoyltransferase 1a (Cpt1a) is considered the rate-limiting enzyme for mitochondrial metabolism of long-chain fatty acids, and Cpt1a deficiency is associated with infant mortality and infection risk. This study was undertaken to test the hypothesis that impairment in Cpt1a-dependent fatty acid oxidation results in increased susceptibility to infection. Screening the Cpt1a gene for common variants predicted to affect protein function revealed allele rs2229738_T, which was associated with pneumonia risk in a targeted human phenome association study. Pharmacologic inhibition of Cpt1a increases mortality and impairs control of the infection in a murine model of bacterial pneumonia. Susceptibility to pneumonia is associated with blunted neutrophilic responses in mice and humans that result from impaired neutrophil trafficking to the site of infection. Chemotaxis responsible for neutrophil trafficking requires Cpt1a-dependent mitochondrial fatty acid oxidation for amplification of chemoattractant signals. These findings identify Cpt1a as a potential host determinant of infection susceptibility and demonstrate a requirement for mitochondrial fatty acid oxidation in neutrophil biology.


Asunto(s)
Carnitina O-Palmitoiltransferasa , Metabolismo de los Lípidos , Neutrófilos , Animales , Humanos , Lactante , Ratones , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Mitocondrias/metabolismo , Neutrófilos/metabolismo
5.
NAR Cancer ; 4(1): zcac007, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35252869

RESUMEN

Rhabdoid tumors (RT) are rare and deadly pediatric cancers driven by loss of SMARCB1, which encodes the SNF5 component of the SWI/SNF chromatin remodeler. Loss of SMARCB1 is associated with a complex set of phenotypic changes including vulnerability to inhibitors of protein synthesis and of the p53 ubiquitin-ligase HDM2. Recently, we discovered small molecule inhibitors of the 'WIN' site of WDR5, which in MLL-rearranged leukemia cells decrease the expression of a set of genes linked to protein synthesis, inducing a translational choke and causing p53-dependent inhibition of proliferation. Here, we characterize how WIN site inhibitors act in RT cells. As in leukemia cells, WIN site inhibition in RT cells causes the comprehensive displacement of WDR5 from chromatin, resulting in a decrease in protein synthesis gene expression. Unlike leukemia cells, however, the growth response of RT cells to WIN site blockade is independent of p53. Exploiting this observation, we demonstrate that WIN site inhibitor synergizes with an HDM2 antagonist to induce p53 and block RT cell proliferation in vitro. These data reveal a p53-independent action of WIN site inhibitors and forecast that future strategies to treat RT could be based on dual WDR5/HDM2 inhibition.

6.
Sci Rep ; 12(1): 1848, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35115608

RESUMEN

WDR5 nucleates the assembly of histone-modifying complexes and acts outside this context in a range of chromatin-centric processes. WDR5 is also a prominent target for pharmacological inhibition in cancer. Small-molecule degraders of WDR5 have been described, but most drug discovery efforts center on blocking the WIN site of WDR5, an arginine binding cavity that engages MLL/SET enzymes that deposit histone H3 lysine 4 methylation (H3K4me). Therapeutic application of WIN site inhibitors is complicated by the disparate functions of WDR5, but is generally guided by two assumptions-that WIN site inhibitors disable all functions of WDR5, and that changes in H3K4me drive the transcriptional response of cancer cells to WIN site blockade. Here, we test these assumptions by comparing the impact of WIN site inhibition versus WDR5 degradation on H3K4me and transcriptional processes. We show that WIN site inhibition disables only a specific subset of WDR5 activity, and that H3K4me changes induced by WDR5 depletion do not explain accompanying transcriptional responses. These data recast WIN site inhibitors as selective loss-of-function agents, contradict H3K4me as a relevant mechanism of action for WDR5 inhibitors, and indicate distinct clinical applications of WIN site inhibitors and WDR5 degraders.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Linfoma de Células B/tratamiento farmacológico , Sitios de Unión , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Metilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Transducción de Señal , Transcripción Genética
7.
Development ; 148(24)2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34927678

RESUMEN

Lung organogenesis requires precise timing and coordination to effect spatial organization and function of the parenchymal cells. To provide a systematic broad-based view of the mechanisms governing the dynamic alterations in parenchymal cells over crucial periods of development, we performed a single-cell RNA-sequencing time-series yielding 102,571 epithelial, endothelial and mesenchymal cells across nine time points from embryonic day 12 to postnatal day 14 in mice. Combining computational fate-likelihood prediction with RNA in situ hybridization and immunofluorescence, we explore lineage relationships during the saccular to alveolar stage transition. The utility of this publicly searchable atlas resource (www.sucrelab.org/lungcells) is exemplified by discoveries of the complexity of type 1 pneumocyte function and characterization of mesenchymal Wnt expression patterns during the saccular and alveolar stages - wherein major expansion of the gas-exchange surface occurs. We provide an integrated view of cellular dynamics in epithelial, endothelial and mesenchymal cell populations during lung organogenesis.


Asunto(s)
Desarrollo Embrionario/genética , Pulmón/crecimiento & desarrollo , Células Madre Mesenquimatosas/citología , Organogénesis/genética , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Embrión de Mamíferos/ultraestructura , Células Epiteliales/citología , Células Epiteliales/ultraestructura , Regulación del Desarrollo de la Expresión Génica/genética , Pulmón/ultraestructura , Células Madre Mesenquimatosas/ultraestructura , Ratones , RNA-Seq , Análisis de la Célula Individual , Transcriptoma/genética
8.
Oncogene ; 40(20): 3593-3609, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33931740

RESUMEN

The SNF5 subunit of the SWI/SNF chromatin remodeling complex has been shown to act as a tumor suppressor through multiple mechanisms, including impairing the ability of the oncoprotein transcription factor MYC to bind chromatin. Beyond SNF5, however, it is unknown to what extent MYC can access additional SWI/SNF subunits or how these interactions affect the ability of MYC to drive transcription, particularly in SNF5-null cancers. Here, we report that MYC interacts with multiple SWI/SNF components independent of SNF5. We show that MYC binds the pan-SWI/SNF subunit BAF155 through the BAF155 SWIRM domain, an interaction that is inhibited by the presence of SNF5. In SNF5-null cells, MYC binds with remaining SWI/SNF components to essential genes, although for a purpose that is distinct from chromatin remodeling. Analysis of MYC-SWI/SNF target genes in SNF5-null cells reveals that they are associated with core biological functions of MYC linked to protein synthesis. These data reveal that MYC can bind SWI/SNF in an SNF5-independent manner and that SNF5 modulates access of MYC to core SWI/SNF complexes. This work provides a framework in which to interrogate the influence of SWI/SNF on MYC function in cancers in which SWI/SNF or MYC are altered.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Neoplasias/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína SMARCB1/metabolismo , Factores de Transcripción/metabolismo , Línea Celular , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/genética , Células HEK293 , Humanos , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteína SMARCB1/genética , Factores de Transcripción/genética
9.
Gastroenterology ; 160(3): 755-770.e26, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33010250

RESUMEN

BACKGROUND & AIMS: The enteric nervous system (ENS) coordinates essential intestinal functions through the concerted action of diverse enteric neurons (ENs). However, integrated molecular knowledge of EN subtypes is lacking. To compare human and mouse ENs, we transcriptionally profiled healthy ENS from adult humans and mice. We aimed to identify transcripts marking discrete neuron subtypes and visualize conserved EN subtypes for humans and mice in multiple bowel regions. METHODS: Human myenteric ganglia and adjacent smooth muscle were isolated by laser-capture microdissection for RNA sequencing. Ganglia-specific transcriptional profiles were identified by computationally subtracting muscle gene signatures. Nuclei from mouse myenteric neurons were isolated and subjected to single-nucleus RNA sequencing, totaling more than 4 billion reads and 25,208 neurons. Neuronal subtypes were defined using mouse single-nucleus RNA sequencing data. Comparative informatics between human and mouse data sets identified shared EN subtype markers, which were visualized in situ using hybridization chain reaction. RESULTS: Several EN subtypes in the duodenum, ileum, and colon are conserved between humans and mice based on orthologous gene expression. However, some EN subtype-specific genes from mice are expressed in completely distinct morphologically defined subtypes in humans. In mice, we identified several neuronal subtypes that stably express gene modules across all intestinal segments, with graded, regional expression of 1 or more marker genes. CONCLUSIONS: Our combined transcriptional profiling of human myenteric ganglia and mouse EN provides a rich foundation for developing novel intestinal therapeutics. There is congruency among some EN subtypes, but we note multiple species differences that should be carefully considered when relating findings from mouse ENS research to human gastrointestinal studies.


Asunto(s)
Diferenciación Celular/genética , Sistema Nervioso Entérico/fisiología , Regulación de la Expresión Génica/fisiología , Neuronas/metabolismo , Especificidad de la Especie , Adolescente , Adulto , Animales , Núcleo Celular/metabolismo , Colon/citología , Colon/inervación , Modelos Animales de Enfermedad , Duodeno/citología , Duodeno/inervación , Femenino , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/fisiopatología , Motilidad Gastrointestinal , Humanos , Íleon/citología , Íleon/inervación , Captura por Microdisección con Láser , Masculino , Ratones , Ratones Transgénicos , Neuronas/citología , RNA-Seq , Factores Sexuales , Análisis de la Célula Individual , Adulto Joven
10.
Nucleic Acids Res ; 48(6): 2924-2941, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-31996893

RESUMEN

WDR5 is a highly-conserved nuclear protein that performs multiple scaffolding functions in the context of chromatin. WDR5 is also a promising target for pharmacological inhibition in cancer, with small molecule inhibitors of an arginine-binding pocket of WDR5 (the 'WIN' site) showing efficacy against a range of cancer cell lines in vitro. Efforts to understand WDR5, or establish the mechanism of action of WIN site inhibitors, however, are stymied by its many functions in the nucleus, and a lack of knowledge of the conserved gene networks-if any-that are under its control. Here, we have performed comparative genomic analyses to identify the conserved sites of WDR5 binding to chromatin, and the conserved genes regulated by WDR5, across a diverse panel of cancer cell lines. We show that a specific cohort of protein synthesis genes (PSGs) are invariantly bound by WDR5, demonstrate that the WIN site anchors WDR5 to chromatin at these sites, and establish that PSGs are bona fide, acute, and persistent targets of WIN site blockade. Together, these data reveal that WDR5 plays a predominant transcriptional role in biomass accumulation and provide further evidence that WIN site inhibitors act to repress gene networks linked to protein synthesis homeostasis.


Asunto(s)
Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Biosíntesis de Proteínas/genética , Secuencia de Bases , Sitios de Unión/genética , Línea Celular , Cromatina/metabolismo , Secuencia Conservada/genética , Femenino , Humanos , Masculino , Unión Proteica , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
11.
Physiol Rep ; 7(23): e14303, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31814333

RESUMEN

There has been a resurgence of interest in the volume-regulated anion channel (VRAC) since the recent cloning of the LRRC8A-E gene family that encodes VRAC. The channel is a heteromer comprised of LRRC8A and at least one other family member; disruption of LRRC8A expression abolishes VRAC activity. The best-in-class VRAC inhibitor, DCPIB, suffers from off-target activity toward several different channels and transporters. Considering that some anion channel inhibitors also suppress mitochondrial respiration, we systematically explored whether DCPIB inhibits respiration in wild type (WT) and LRRC8A-knockout HAP-1 and HEK-293 cells. Knockout of LRRC8A had no apparent effects on cell morphology, proliferation rate, mitochondrial content, or expression of several mitochondrial genes in HAP-1 cells. Addition of 10 µM DCPIB, a concentration typically used to inhibit VRAC, suppressed basal and ATP-linked respiration in part through uncoupling the inner mitochondrial membrane (IMM) proton gradient and membrane potential. Additionally, DCPIB inhibits the activity of complex I, II, and III of the electron transport chain (ETC). Surprisingly, the effects of DCPIB on mitochondrial function are also observed in HAP-1 and HEK-293 cells which lack LRRC8A expression. Finally, we demonstrate that DCPIB activates ATP-inhibitable potassium channels comprised of heterologously expressed Kir6.2 and SUR1 subunits. These data indicate that DCPIB suppresses mitochondrial respiration and ATP production by dissipating the mitochondrial membrane potential and inhibiting complexes I-III of the ETC. They further justify the need for the development of sharper pharmacological tools for evaluating the integrative physiology and therapeutic potential of VRAC in human diseases.


Asunto(s)
Ciclopentanos/farmacología , Indanos/farmacología , Proteínas de la Membrana/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Línea Celular Tumoral , Respiración de la Célula/efectos de los fármacos , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Células HEK293 , Humanos , Canales KATP/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo
12.
Am J Physiol Renal Physiol ; 316(5): F847-F855, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30759021

RESUMEN

Flow cytometry studies on injured kidney tubules are complicated by the low yield of nucleated single cells. Furthermore, cell-specific responses such as cell cycle dynamics in vivo have conventionally relied on indirect immunohistochemistry and proximal tubule markers that may be downregulated in injury. Here, we report a new tissue dissociation protocol for the kidney with an early fixation step that greatly enhances the yield of single cells. Genetic labeling of the proximal tubule with either mT/mG "tomato" or R26Fucci2aR (Fucci) cell cycle reporter mice allows us to follow proximal tubule-specific changes in cell cycle after renal injury. Image-based flow cytometry (FlowSight) enables gating of the cell cycle and concurrent visualization of the cells with bright field and fluorescence. We used the Fucci mouse in conjunction with FlowSight to identify a discrete polyploid population in proximal tubules after aristolochic acid injury. The tissue dissociation protocol in conjunction with genetic labeling and image-based flow cytometry is a tool that can improve our understanding of any discrete cell population after injury.


Asunto(s)
Lesión Renal Aguda/patología , Ciclo Celular , Separación Celular/métodos , Células Epiteliales/patología , Citometría de Flujo , Túbulos Renales Proximales/patología , Fijación del Tejido/métodos , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Animales , Ácidos Aristolóquicos , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Genes Reporteros , Túbulos Renales Proximales/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones Transgénicos , Poliploidía
13.
Int J Nanomedicine ; 11: 6103-6121, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27895483

RESUMEN

The present study investigated the immunoenhancing property of our newly designed nanovaccine, that is, its ability to induce antigen-specific immunity. This study also evaluated the synergistic effect of a novel compound PBS-44, an α-galactosylceramide analog, in boosting the immune response induced by our nanovaccine. The nanovaccine was prepared by encapsulating ovalbumin (ova) and an adjuvant within the poly(lactic-co-glycolic acid) nanoparticles. Quantitative analysis of our study data showed that the encapsulated vaccine was physically and biologically stable; the core content of our nanovaccine was found to be released steadily and slowly, and nearly 90% of the core content was slowly released over the course of 25 days. The in vivo immunization studies exhibited that the nanovaccine induced stronger and longer immune responses compared to its soluble counterpart. Similarly, intranasal inhalation of the nanovaccine induced more robust antigen-specific CD8+ T cell response than intraperitoneal injection of nanovaccine.


Asunto(s)
Galactosilceramidas/farmacología , Nanopartículas , Linfocitos T/efectos de los fármacos , Vacunas/administración & dosificación , Vacunas/farmacología , Adyuvantes Inmunológicos/farmacología , Administración Intranasal , Animales , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Dendríticas/inmunología , Galactosilceramidas/química , Inmunización , Inyecciones Intraperitoneales , Ácido Láctico/química , Ratones , Ratones Endogámicos C57BL , Microscopía de Fuerza Atómica , Nanopartículas/química , Ovalbúmina/química , Ovalbúmina/inmunología , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Linfocitos T/inmunología , Vacunas/química , Vacunas/inmunología
14.
Cytometry A ; 89(3): 271-80, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26599989

RESUMEN

Mass and fluorescence cytometry are quantitative single cell flow cytometry approaches that are powerful tools for characterizing diverse tissues and cellular systems. Here mass cytometry was directly compared with fluorescence cytometry by studying phenotypes of healthy human peripheral blood mononuclear cells (PBMC) in the context of superantigen stimulation. One mass cytometry panel and five fluorescence cytometry panels were used to measure 20 well-established lymphocyte markers of memory and activation. Comparable frequencies of both common and rare cell subpopulations were observed with fluorescence and mass cytometry using biaxial gating. The unsupervised high-dimensional analysis tool viSNE was then used to analyze data sets generated from both mass and fluorescence cytometry. viSNE analysis effectively characterized PBMC using eight features per cell and identified similar frequencies of activated CD4+ T cells with both technologies. These results suggest combinations of unsupervised analysis programs and extended multiparameter cytometry will be indispensable tools for detecting perturbations in protein expression in both health and disease.


Asunto(s)
Citometría de Flujo/normas , Inmunofenotipificación/métodos , Leucocitos Mononucleares/citología , Espectrometría de Masas/normas , Antígenos CD/genética , Antígenos CD/inmunología , Expresión Génica , Humanos , Elementos de la Serie de los Lantanoides/análisis , Leucocitos Mononucleares/clasificación , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Espectrometría de Masas/instrumentación , Espectrometría de Masas/métodos , Análisis Multivariante
15.
Arthritis Rheumatol ; 66(11): 2947-57, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25077978

RESUMEN

OBJECTIVE: To determine interrelationships between the expression of long intergenic (noncoding) RNA-p21 (lincRNA-p21), NF-κB activity, and responses to methotrexate (MTX) in rheumatoid arthritis (RA) by analyzing patient blood samples and cell culture models. METHODS: Expression levels of long noncoding RNA and messenger RNA (mRNA) were determined by quantitative reverse transcription-polymerase chain reaction. Western blotting and flow cytometry were used to quantify levels of intracellular proteins. Intracellular NF-κB activity was determined using an NF-κB luciferase reporter plasmid. RESULTS: Patients with RA expressed reduced basal levels of lincRNA-p21 and increased basal levels of phosphorylated p65 (RelA), a marker of NF-κB activation. Patients with RA who were not treated with MTX expressed lower levels of lincRNA-p21 and higher levels of phosphorylated p65 compared with RA patients treated with low-dose MTX. In cell culture using primary cells and transformed cell lines, MTX induced lincRNA-p21 through a DNA-dependent protein kinase catalytic subunit (DNA PKcs)-dependent mechanism. Deficiencies in the levels of PRKDC mRNA in patients with RA were also corrected by MTX in vivo. Furthermore, MTX reduced NF-κB activity in tumor necrosis factor α-treated cells through a DNA PKcs-dependent mechanism via induction of lincRNA-p21. Finally, we observed that depressed levels of TP53 and lincRNA-p21 increased NF-κB activity in cell lines. Decreased levels of lincRNA-p21 did not alter NFKB1 or RELA transcripts; rather, lincRNA-p21 physically bound to RELA mRNA. CONCLUSION: Our findings support a model whereby depressed levels of lincRNA-p21 in RA contribute to increased NF-κB activity. MTX decreases basal levels of NF-κB activity by increasing lincRNA-p21 levels through a DNA PKcs-dependent mechanism.


Asunto(s)
Antirreumáticos/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Metotrexato/farmacología , FN-kappa B/metabolismo , ARN Largo no Codificante/metabolismo , Adulto , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Línea Celular , Células Cultivadas , Proteína Quinasa Activada por ADN/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Metotrexato/uso terapéutico , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/patología , Proteínas Nucleares/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T/patología , Proteína p53 Supresora de Tumor/metabolismo , eIF-2 Quinasa/metabolismo
16.
Brain Behav Immun ; 35: 33-42, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23831150

RESUMEN

Obesity is associated with chronic low-grade inflammation in peripheral tissues caused, in part, by the recruitment of inflammatory monocytes into adipose tissue. Studies in rodent models have also shown increased inflammation in the central nervous system (CNS) during obesity. The goal of this study was to determine whether obesity is associated with recruitment of peripheral immune cells into the CNS. To do this we used a bone marrow chimerism model to track the entry of green-fluorescent protein (GFP) labeled peripheral immune cells into the CNS. Flow cytometry was used to quantify the number of GFP(+) immune cells recruited into the CNS of mice fed a high-fat diet compared to standard chow fed controls. High-fat feeding resulted in obesity associated with a 30% increase in the number of GFP(+) cells in the CNS compared to control mice. Greater than 80% of the GFP(+) cells recruited to the CNS were also CD45(+) CD11b(+) indicating that the GFP(+) cells displayed characteristics of microglia/macrophages. Immunohistochemistry further confirmed the increase in GFP(+) cells in the CNS of the high-fat fed group and also indicated that 93% of the recruited cells were found in the parenchyma and had a stellate morphology. These findings indicate that peripheral immune cells can be recruited to the CNS in obesity and may contribute to the inflammatory response.


Asunto(s)
Encéfalo/inmunología , Dieta Alta en Grasa/efectos adversos , Monocitos/metabolismo , Neuroinmunomodulación , Obesidad/inmunología , Animales , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo
18.
Stem Cell Reports ; 1(6): 532-44, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24371808

RESUMEN

Heterogeneity within pluripotent stem cell (PSC) populations is indicative of dynamic changes that occur when cells drift between different states. Although the role of metastability in PSCs is unclear, it appears to reflect heterogeneity in cell signaling. Using the Fucci cell-cycle indicator system, we show that elevated expression of developmental regulators in G1 is a major determinant of heterogeneity in human embryonic stem cells. Although signaling pathways remain active throughout the cell cycle, their contribution to heterogeneous gene expression is restricted to G1. Surprisingly, we identify dramatic changes in the levels of global 5-hydroxymethylcytosine, an unanticipated source of epigenetic heterogeneity that is tightly linked to cell-cycle progression and the expression of developmental regulators. When we evaluated gene expression in differentiating cells, we found that cell-cycle regulation of developmental regulators was maintained during lineage specification. Cell-cycle regulation of developmentally regulated transcription factors is therefore an inherent feature of the mechanisms underpinning differentiation.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes/citología , 5-Metilcitosina/análogos & derivados , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Citosina/análogos & derivados , Citosina/metabolismo , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA