Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 182(6): 1460-1473.e17, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32916129

RESUMEN

The gut microbiome has been implicated in multiple human chronic gastrointestinal (GI) disorders. Determining its mechanistic role in disease has been difficult due to apparent disconnects between animal and human studies and lack of an integrated multi-omics view of disease-specific physiological changes. We integrated longitudinal multi-omics data from the gut microbiome, metabolome, host epigenome, and transcriptome in the context of irritable bowel syndrome (IBS) host physiology. We identified IBS subtype-specific and symptom-related variation in microbial composition and function. A subset of identified changes in microbial metabolites correspond to host physiological mechanisms that are relevant to IBS. By integrating multiple data layers, we identified purine metabolism as a novel host-microbial metabolic pathway in IBS with translational potential. Our study highlights the importance of longitudinal sampling and integrating complementary multi-omics data to identify functional mechanisms that can serve as therapeutic targets in a comprehensive treatment strategy for chronic GI diseases. VIDEO ABSTRACT.


Asunto(s)
Microbioma Gastrointestinal/genética , Regulación de la Expresión Génica/genética , Síndrome del Colon Irritable/metabolismo , Metaboloma , Purinas/metabolismo , Transcriptoma/genética , Animales , Ácidos y Sales Biliares/metabolismo , Biopsia , Butiratos/metabolismo , Cromatografía Liquida , Estudios Transversales , Epigenómica , Heces/microbiología , Femenino , Microbioma Gastrointestinal/fisiología , Regulación de la Expresión Génica/fisiología , Interacciones Microbiota-Huesped/genética , Humanos , Hipoxantina/metabolismo , Síndrome del Colon Irritable/genética , Síndrome del Colon Irritable/microbiología , Estudios Longitudinales , Masculino , Metaboloma/fisiología , Ratones , Estudios Observacionales como Asunto , Estudios Prospectivos , Programas Informáticos , Espectrometría de Masas en Tándem , Transcriptoma/fisiología
3.
Mol Cell ; 65(2): 272-284, 2017 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-28107649

RESUMEN

The histone chaperone HIRA is involved in depositing histone variant H3.3 into distinct genic regions, including promoters, enhancers, and gene bodies. However, how HIRA deposits H3.3 to these regions remains elusive. Through a short hairpin RNA (shRNA) screening, we identified single-stranded DNA binding protein replication protein A (RPA) as a regulator of the deposition of newly synthesized H3.3 into chromatin. We show that RPA physically interacts with HIRA to form RPA-HIRA-H3.3 complexes, and it co-localizes with HIRA and H3.3 at gene promoters and enhancers. Depletion of RPA1, the largest subunit of the RPA complex, dramatically reduces both HIRA association with chromatin and the deposition of newly synthesized H3.3 at promoters and enhancers and leads to altered transcription at gene promoters. These results support a model whereby RPA, best known for its role in DNA replication and repair, recruits HIRA to promoters and enhancers and regulates deposition of newly synthesized H3.3 to these regulatory elements for gene regulation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Elementos de Facilitación Genéticos , Chaperonas de Histonas/metabolismo , Histonas/metabolismo , Regiones Promotoras Genéticas , Proteína de Replicación A/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Sitios de Unión , Proteínas de Ciclo Celular/genética , Cromatina/genética , ADN/genética , Proteínas de Unión al ADN/genética , Fase G1 , Células HEK293 , Células HeLa , Chaperonas de Histonas/genética , Humanos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , Proteína de Replicación A/genética , Factores de Transcripción/genética , Transfección
4.
Gastroenterology ; 165(6): 1458-1474, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37597632

RESUMEN

BACKGROUND & AIMS: Although depletion of neuronal nitric oxide synthase (NOS1)-expressing neurons contributes to gastroparesis, stimulating nitrergic signaling is not an effective therapy. We investigated whether hypoxia-inducible factor 1α (HIF1A), which is activated by high O2 consumption in central neurons, is a Nos1 transcription factor in enteric neurons and whether stabilizing HIF1A reverses gastroparesis. METHODS: Mice with streptozotocin-induced diabetes, human and mouse tissues, NOS1+ mouse neuroblastoma cells, and isolated nitrergic neurons were studied. Gastric emptying of solids and volumes were determined by breath test and single-photon emission computed tomography, respectively. Gene expression was analyzed by RNA-sequencing, microarrays, immunoblotting, and immunofluorescence. Epigenetic assays included chromatin immunoprecipitation sequencing (13 targets), chromosome conformation capture sequencing, and reporter assays. Mechanistic studies used Cre-mediated recombination, RNA interference, and clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9)-mediated epigenome editing. RESULTS: HIF1A signaling from physiological intracellular hypoxia was active in mouse and human NOS1+ myenteric neurons but reduced in diabetes. Deleting Hif1a in Nos1-expressing neurons reduced NOS1 protein by 50% to 92% and delayed gastric emptying of solids in female but not male mice. Stabilizing HIF1A with roxadustat (FG-4592), which is approved for human use, restored NOS1 and reversed gastroparesis in female diabetic mice. In nitrergic neurons, HIF1A up-regulated Nos1 transcription by binding and activating proximal and distal cis-regulatory elements, including newly discovered super-enhancers, facilitating RNA polymerase loading and pause-release, and by recruiting cohesin to loop anchors to alter chromosome topology. CONCLUSIONS: Pharmacologic HIF1A stabilization is a novel, translatable approach to restoring nitrergic signaling and treating diabetic gastroparesis. The newly recognized effects of HIF1A on chromosome topology may provide insights into physioxia- and ischemia-related organ function.


Asunto(s)
Diabetes Mellitus Experimental , Gastroparesia , Animales , Femenino , Humanos , Ratones , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Epigénesis Genética , Gastroparesia/genética , Neuronas , Óxido Nítrico Sintasa de Tipo I
5.
Nucleic Acids Res ; 50(20): 11635-11653, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36399508

RESUMEN

Understanding the function of non-coding genomic sequence variants represents a challenge for biomedicine. Many diseases are products of gene-by-environment interactions with complex mechanisms. This study addresses these themes by mechanistic characterization of non-coding variants that influence gene expression only after drug or hormone exposure. Using glucocorticoid signaling as a model system, we integrated genomic, transcriptomic, and epigenomic approaches to unravel mechanisms by which variant function could be revealed by hormones or drugs. Specifically, we identified cis-regulatory elements and 3D interactions underlying ligand-dependent associations between variants and gene expression. One-quarter of the glucocorticoid-modulated variants that we identified had already been associated with clinical phenotypes. However, their affected genes were 'unmasked' only after glucocorticoid exposure and often with function relevant to the disease phenotypes. These diseases involved glucocorticoids as risk factors or therapeutic agents and included autoimmunity, metabolic and mood disorders, osteoporosis and cancer. For example, we identified a novel breast cancer risk gene, MAST4, with expression that was repressed by glucocorticoids in cells carrying the risk genotype, repression that correlated with MAST4 expression in breast cancer and treatment outcomes. These observations provide a mechanistic framework for understanding non-coding genetic variant-chemical environment interactions and their role in disease risk and drug response.


Asunto(s)
Glucocorticoides , Secuencias Reguladoras de Ácidos Nucleicos , Glucocorticoides/genética , Glucocorticoides/metabolismo , Factores de Riesgo , Humanos , Farmacogenética , Sitios de Carácter Cuantitativo
6.
Genes Dev ; 30(8): 946-59, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-27083998

RESUMEN

Dynamic regulation of RNF168-mediated ubiquitylation of histone H2A Lys13,15 (H2AK13,15ub) at DNA double-strand breaks (DSBs) is crucial for preventing aberrant DNA repair and maintaining genome stability. However, it remains unclear which deubiquitylating enzyme (DUB) removes H2AK13,15ub. Here we show that USP51, a previously uncharacterized DUB, deubiquitylates H2AK13,15ub and regulates DNA damage response. USP51 depletion results in increased spontaneous DNA damage foci and elevated levels of H2AK15ub and impairs DNA damage response. USP51 overexpression suppresses the formation of ionizing radiation-induced 53BP1 and BRCA1 but not RNF168 foci, suggesting that USP51 functions downstream from RNF168 in DNA damage response. In vitro, USP51 binds to H2A-H2B directly and deubiquitylates H2AK13,15ub. In cells, USP51 is recruited to chromatin after DNA damage and regulates the dynamic assembly/disassembly of 53BP1 and BRCA1 foci. These results show that USP51 is the DUB for H2AK13,15ub and regulates DNA damage response.


Asunto(s)
Daño del ADN/fisiología , Reparación del ADN/fisiología , Histonas/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular/fisiología , ADN/metabolismo , ADN/efectos de la radiación , Humanos , Proteínas Nucleares/metabolismo , Unión Proteica , Radiación Ionizante , Transactivadores/metabolismo , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación
7.
BMC Genomics ; 24(1): 171, 2023 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-37016279

RESUMEN

Chromatin immunoprecipitation (ChIP) is an antibody-based approach that is frequently utilized in chromatin biology and epigenetics. The challenge in experimental variability by unpredictable nature of usable input amounts from samples and undefined antibody titer in ChIP reaction still remains to be addressed. Here, we introduce a simple and quick method to quantify chromatin inputs and demonstrate its utility for normalizing antibody amounts to the optimal titer in individual ChIP reactions. For a proof of concept, we utilized ChIP-seq validated antibodies against the key enhancer mark, acetylation of histone H3 on lysine 27 (H3K27ac), in the experiments. The results indicate that the titration-based normalization of antibody amounts improves assay outcomes including the consistency among samples both within and across experiments for a broad range of input amounts.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina , Histonas , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Inmunoprecipitación de Cromatina/métodos , Histonas/genética , Cromatina , Anticuerpos
8.
Gastroenterology ; 162(2): 535-547.e13, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34688712

RESUMEN

BACKGROUND AND AIMS: The gastrointestinal (GI) tract extracts nutrients from ingested meals while protecting the organism from infectious agents frequently present in meals. Consequently, most animals conduct the entire digestive process within the GI tract while keeping the luminal contents entirely outside the body, separated by the tightly sealed GI epithelium. Therefore, like the skin and oral cavity, the GI tract must sense the chemical and physical properties of the its external interface to optimize its function. Specialized sensory enteroendocrine cells (EECs) in GI epithelium interact intimately with luminal contents. A subpopulation of EECs express the mechanically gated ion channel Piezo2 and are developmentally and functionally like the skin's touch sensor- the Merkel cell. We hypothesized that Piezo2+ EECs endow the gut with intrinsic tactile sensitivity. METHODS: We generated transgenic mouse models with optogenetic activators in EECs and Piezo2 conditional knockouts. We used a range of reference standard and novel techniques from single cells to living animals, including single-cell RNA sequencing and opto-electrophysiology, opto-organ baths with luminal shear forces, and in vivo studies that assayed GI transit while manipulating the physical properties of luminal contents. RESULTS: Piezo2+ EECs have transcriptomic features of synaptically connected, mechanosensory epithelial cells. EEC activation by optogenetics and forces led to Piezo2-dependent alterations in colonic propagating contractions driven by intrinsic circuitry, with Piezo2+ EECs detecting the small luminal forces and physical properties of the luminal contents to regulate transit times in the small and large bowel. CONCLUSIONS: The GI tract has intrinsic tactile sensitivity that depends on Piezo2+ EECs and allows it to detect luminal forces and physical properties of luminal contents to modulate physiology.


Asunto(s)
Células Enteroendocrinas/metabolismo , Mucosa Intestinal/metabolismo , Canales Iónicos/genética , Tacto/fisiología , Animales , Células Enteroendocrinas/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Técnicas de Inactivación de Genes , Mucosa Intestinal/citología , Mucosa Intestinal/fisiología , Canales Iónicos/metabolismo , Mecanorreceptores , Ratones , Ratones Transgénicos , Optogenética , Peristaltismo/fisiología
9.
Mol Psychiatry ; 26(12): 7454-7464, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34535768

RESUMEN

Bipolar disorder (BD) and obesity are highly comorbid. We previously performed a genome-wide association study (GWAS) for BD risk accounting for the effect of body mass index (BMI), which identified a genome-wide significant single-nucleotide polymorphism (SNP) in the gene encoding the transcription factor 7 like 2 (TCF7L2). However, the molecular function of TCF7L2 in the central nervous system (CNS) and its possible role in the BD and BMI interaction remained unclear. In the present study, we demonstrated by studying human induced pluripotent stem cell (hiPSC)-derived astrocytes, cells that highly express TCF7L2 in the CNS, that the BD-BMI GWAS risk SNP is associated with glucocorticoid-dependent repression of the expression of a previously uncharacterized TCF7L2 transcript variant. That transcript is a long non-coding RNA (lncRNA-TCF7L2) that is highly expressed in the CNS but not in peripheral tissues such as the liver and pancreas that are involved in metabolism. In astrocytes, knockdown of the lncRNA-TCF7L2 resulted in decreased expression of the parent gene, TCF7L2, as well as alterations in the expression of a series of genes involved in insulin signaling and diabetes. We also studied the function of TCF7L2 in hiPSC-derived astrocytes by integrating RNA sequencing data after TCF7L2 knockdown with TCF7L2 chromatin-immunoprecipitation sequencing (ChIP-seq) data. Those studies showed that TCF7L2 directly regulated a series of BD risk genes. In summary, these results support the existence of a CNS-based mechanism underlying BD-BMI genetic risk, a mechanism based on a glucocorticoid-dependent expression quantitative trait locus that regulates the expression of a novel TCF7L2 non-coding transcript.


Asunto(s)
Trastorno Bipolar , Diabetes Mellitus Tipo 2 , Células Madre Pluripotentes Inducidas , ARN Largo no Codificante , Trastorno Bipolar/genética , Índice de Masa Corporal , Diabetes Mellitus Tipo 2/metabolismo , Estudio de Asociación del Genoma Completo , Glucocorticoides , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Polimorfismo de Nucleótido Simple/genética , ARN Largo no Codificante/genética , Proteína 2 Similar al Factor de Transcripción 7/genética , Proteína 2 Similar al Factor de Transcripción 7/metabolismo
10.
Mol Cell ; 56(4): 551-63, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25449133

RESUMEN

In eukaryotic cells, DNA replication proceeds with continuous synthesis of leading-strand DNA and discontinuous synthesis of lagging-strand DNA. Here we describe a method, eSPAN (enrichment and sequencing of protein-associated nascent DNA), which reveals the genome-wide association of proteins with leading and lagging strands of DNA replication forks. Using this approach in budding yeast, we confirm the strand specificities of DNA polymerases delta and epsilon and show that the PCNA clamp is enriched at lagging strands compared with leading-strand replication. Surprisingly, at stalled forks, PCNA is unloaded specifically from lagging strands. PCNA unloading depends on the Elg1-containing alternative RFC complex, ubiquitination of PCNA, and the checkpoint kinases Mec1 and Rad53. Cells deficient in PCNA unloading exhibit increased chromosome breaks. Our studies provide a tool for studying replication-related processes and reveal a mechanism whereby checkpoint kinases regulate strand-specific unloading of PCNA from stalled replication forks to maintain genome stability.


Asunto(s)
Replicación del ADN , ADN de Hongos/biosíntesis , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Inmunoprecipitación de Cromatina , Cromosomas Fúngicos/genética , Daño del ADN , ADN Polimerasa II/metabolismo , ADN Polimerasa III/metabolismo , ADN de Hongos/genética , Inestabilidad Genómica , Unión Proteica , Análisis de Secuencia de ADN , Ubiquitinación
11.
J Cell Sci ; 132(20)2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31527052

RESUMEN

Tissue fibrosis is a chronic disease driven by persistent fibroblast activation that has recently been linked to epigenetic modifications. Here, we screened a small library of epigenetic small-molecule modulators to identify compounds capable of inhibiting or reversing TGFß-mediated fibroblast activation. We identified pracinostat, an HDAC inhibitor, as a potent attenuator of lung fibroblast activation and confirmed its efficacy in patient-derived fibroblasts isolated from fibrotic lung tissue. Mechanistically, we found that HDAC-dependent transcriptional repression was an early and essential event in TGFß-mediated fibroblast activation. Treatment of lung fibroblasts with pracinostat broadly attenuated TGFß-mediated epigenetic repression and promoted fibroblast quiescence. We confirmed a specific role for HDAC-dependent histone deacetylation in the promoter region of the anti-fibrotic gene PPARGC1A (PGC1α) in response to TGFß stimulation. Finally, we identified HDAC7 as a key factor whose siRNA-mediated knockdown attenuates fibroblast activation without altering global histone acetylation. Together, these results provide novel mechanistic insight into the essential role HDACs play in TGFß-mediated fibroblast activation via targeted gene repression.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Fibroblastos/enzimología , Histona Desacetilasas/metabolismo , Pulmón/enzimología , Fibrosis Pulmonar/enzimología , Factor de Crecimiento Transformador beta/farmacología , Línea Celular , Fibroblastos/patología , Histona Desacetilasas/genética , Humanos , Pulmón/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/biosíntesis , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Regiones Promotoras Genéticas , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología
12.
Blood ; 133(26): 2776-2789, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31101622

RESUMEN

Anaplastic large cell lymphomas (ALCLs) represent a relatively common group of T-cell non-Hodgkin lymphomas (T-NHLs) that are unified by similar pathologic features but demonstrate marked genetic heterogeneity. ALCLs are broadly classified as being anaplastic lymphoma kinase (ALK)+ or ALK-, based on the presence or absence of ALK rearrangements. Exome sequencing of 62 T-NHLs identified a previously unreported recurrent mutation in the musculin gene, MSC E116K, exclusively in ALK- ALCLs. Additional sequencing for a total of 238 T-NHLs confirmed the specificity of MSC E116K for ALK- ALCL and further demonstrated that 14 of 15 mutated cases (93%) had coexisting DUSP22 rearrangements. Musculin is a basic helix-loop-helix (bHLH) transcription factor that heterodimerizes with other bHLH proteins to regulate lymphocyte development. The E116K mutation localized to the DNA binding domain of musculin and permitted formation of musculin-bHLH heterodimers but prevented their binding to authentic target sequence. Functional analysis showed MSCE116K acted in a dominant-negative fashion, reversing wild-type musculin-induced repression of MYC and cell cycle inhibition. Chromatin immunoprecipitation-sequencing and transcriptome analysis identified the cell cycle regulatory gene E2F2 as a direct transcriptional target of musculin. MSCE116K reversed E2F2-induced cell cycle arrest and promoted expression of the CD30-IRF4-MYC axis, whereas its expression was reciprocally induced by binding of IRF4 to the MSC promoter. Finally, ALCL cells expressing MSC E116K were preferentially targeted by the BET inhibitor JQ1. These findings identify a novel recurrent MSC mutation as a key driver of the CD30-IRF4-MYC axis and cell cycle progression in a unique subset of ALCLs.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Linfoma Anaplásico de Células Grandes/genética , Quinasa de Linfoma Anaplásico/genética , Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Mutación
13.
Nucleic Acids Res ; 47(21): 11114-11131, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31586391

RESUMEN

Establishment and subsequent maintenance of distinct chromatin domains during embryonic stem cell (ESC) differentiation are crucial for lineage specification and cell fate determination. Here we show that the histone chaperone Chromatin Assembly Factor 1 (CAF-1), which is recruited to DNA replication forks through its interaction with proliferating cell nuclear antigen (PCNA) for nucleosome assembly, participates in the establishment of H3K27me3-mediated silencing during differentiation. Deletion of CAF-1 p150 subunit impairs the silencing of many genes including Oct4, Sox2 and Nanog as well as the establishment of H3K27me3 at these gene promoters during ESC differentiation. Mutations of PCNA residues involved in recruiting CAF-1 to the chromatin also result in defects in differentiation in vitro and impair early embryonic development as p150 deletion. Together, these results reveal that the CAF-1-PCNA nucleosome assembly pathway plays an important role in the establishment of H3K27me3-mediated silencing during cell fate determination.


Asunto(s)
Diferenciación Celular/genética , Factor 1 de Ensamblaje de la Cromatina/fisiología , Células Madre Embrionarias/fisiología , Heterocromatina/metabolismo , Células Madre Pluripotentes/fisiología , Animales , Células Cultivadas , Factor 1 de Ensamblaje de la Cromatina/genética , Factor 1 de Ensamblaje de la Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/genética , Replicación del ADN/genética , Silenciador del Gen/fisiología , Histona Metiltransferasas/metabolismo , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Antígeno Nuclear de Célula en Proliferación/metabolismo
14.
Gut ; 69(5): 868-876, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31757880

RESUMEN

OBJECTIVE: This study was designed to evaluate the roles of microRNAs (miRNAs) in slow transit constipation (STC). DESIGN: All human tissue samples were from the muscularis externa of the colon. Expression of 372 miRNAs was examined in a discovery cohort of four patients with STC versus three age/sex-matched controls by a quantitative PCR array. Upregulated miRNAs were examined by quantitative reverse transcription qPCR (RT-qPCR) in a validation cohort of seven patients with STC and age/sex-matched controls. The effect of a highly differentially expressed miRNA on a custom human smooth muscle cell line was examined in vitro by RT-qPCR, electrophysiology, traction force microscopy, and ex vivo by lentiviral transduction in rat muscularis externa organotypic cultures. RESULTS: The expression of 13 miRNAs was increased in STC samples. Of those miRNAs, four were predicted to target SCN5A, the gene that encodes the Na+ channel NaV1.5. The expression of SCN5A mRNA was decreased in STC samples. Let-7f significantly decreased Na+ current density in vitro in human smooth muscle cells. In rat muscularis externa organotypic cultures, overexpression of let-7f resulted in reduced frequency and amplitude of contraction. CONCLUSIONS: A small group of miRNAs is upregulated in STC, and many of these miRNAs target the SCN5A-encoded Na+ channel NaV1.5. Within this set, a novel NaV1.5 regulator, let-7f, resulted in decreased NaV1.5 expression, current density and reduced motility of GI smooth muscle. These results suggest NaV1.5 and miRNAs as novel diagnostic and potential therapeutic targets in STC.


Asunto(s)
Estreñimiento/fisiopatología , Regulación de la Expresión Génica , MicroARNs/genética , Proteínas Asociadas a Microtúbulos/genética , Contracción Muscular/genética , Adulto , Anciano , Biopsia con Aguja , Estudios de Casos y Controles , Colon/patología , Femenino , Motilidad Gastrointestinal/genética , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Contracción Muscular/fisiología , Músculo Liso , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Valores de Referencia , Muestreo , Regulación hacia Arriba
15.
Anal Chem ; 92(12): 7989-7997, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32496751

RESUMEN

Personalizing health care by taking genetic, environmental, and lifestyle factors into account is central to modern medicine. The crucial and pervasive roles epigenetic factors play in shaping gene-environment interactions are now well recognized. However, identifying robust epigenetic biomarkers and translating them to clinical tests has been difficult due in part to limitations of available platforms to detect epigenetic features genome-wide (epigenomic assays). This Feature introduces several important prospects for precision epigenomics, highlights capabilities and limitations of current laboratory technologies, and emphasizes opportunities for microfluidic tools to facilitate translation of epigenetic analyses to the clinic, with a particular focus on methods to profile gene-associated histone modifications and their impacts on chromatin structure and gene expression.


Asunto(s)
Cromatina/genética , Epigénesis Genética/genética , Epigenómica , Código de Histonas/genética , Dispositivos Laboratorio en un Chip , Humanos
16.
FASEB J ; 33(5): 6632-6642, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30802137

RESUMEN

The Ca2+-activated Cl- channel, anoctamin 1 (Ano1, also known as transmembrane protein 16A) contributes to intestinal pacemaking, fluid secretion, cellular excitability, and tissue development. The human ANO1 promoter contains binding sites for the glioma-associated oncogene (Gli) proteins. We investigated regulation of ANO1 transcription by Gli. ANO1 promoter activity was determined using a luciferase reporter system. Binding and functional effects of Glis on ANO1 transcription and expression were demonstrated by chromatin immunoprecipitation, small interfering RNA knockdown, PCR, immunolabeling, and recordings of Ca2+-activated Cl- currents in human embryonic kidney 293 (HEK293) cells. Results from previous genome-wide association studies were used to test ANO1 promoter polymorphisms for association with disease. Gli1 and Gli2 bound to the promoter and repressed ANO1 transcription. Repression depended on Gli binding to a site close to the ANO1 transcriptional start site. Mutation of this site prevented Gli binding and transcriptional repression. Knockdown of Gli expression and inhibition of Gli activity increased expression of ANO1 RNA and Ca2+-activated Cl- currents in HEK293 cells. A single-nucleotide polymorphism prevented Gli binding and showed association with irritable bowel syndrome. We conclude that Gli1 and Gli2 repress ANO1 by a novel mechanism that is independent of Gli cleavage and that has a role in gastrointestinal function.-Mazzone, A., Gibbons, S. J., Eisenman, S. T., Strege, P. R., Zheng, T., D'Amato, M., Ordog, T., Fernandez-Zapico, M. E., Farrugia, G. Direct repression of anoctamin 1 (ANO1) gene transcription by Gli proteins.


Asunto(s)
Anoctamina-1/biosíntesis , Regulación de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Proteínas Nucleares/metabolismo , Sitio de Iniciación de la Transcripción , Transcripción Genética , Proteína con Dedos de Zinc GLI1/metabolismo , Proteína Gli2 con Dedos de Zinc/metabolismo , Anoctamina-1/genética , Calcio/metabolismo , Señalización del Calcio , Células HEK293 , Humanos , Síndrome del Colon Irritable/genética , Síndrome del Colon Irritable/metabolismo , Mutación , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Polimorfismo de Nucleótido Simple , Proteína con Dedos de Zinc GLI1/genética , Proteína Gli2 con Dedos de Zinc/genética
17.
Proc Natl Acad Sci U S A ; 114(15): 3933-3938, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28348210

RESUMEN

Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase involved in development and human disease, including cancer. It is currently thought that the four-point one, ezrin, radixin, moesin (FERM)-kinase domain linker, which contains autophosphorylation site tyrosine (Y) 397, is not required for in vivo FAK function until late midgestation. Here, we directly tested this hypothesis by generating mice with FAK Y397-to-phenylalanine (F) mutations in the germline. We found that Y397F embryos exhibited reduced mesodermal fibronectin (FN) and osteopontin expression and died during mesoderm development akin to FAK kinase-dead mice. We identified myosin-1E (MYO1E), an actin-dependent molecular motor, to interact directly with the FAK FERM-kinase linker and induce FAK kinase activity and Y397 phosphorylation. Active FAK in turn accumulated in the nucleus where it led to the expression of osteopontin and other FN-type matrix in both mouse embryonic fibroblasts and human melanoma. Our data support a model in which FAK Y397 autophosphorylation is required for FAK function in vivo and is positively regulated by MYO1E.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Melanoma/metabolismo , Miosinas/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Pérdida del Embrión/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/genética , Humanos , Melanoma/patología , Mesodermo/embriología , Ratones Mutantes , Miosina Tipo I , Miosinas/química , Miosinas/genética , Osteopontina/genética , Osteopontina/metabolismo , Fosforilación , Embarazo , Dominios Proteicos , Neoplasias Cutáneas/patología , Tirosina/metabolismo
18.
BMC Genomics ; 20(1): 249, 2019 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-30922218

RESUMEN

BACKGROUND: Epigenetic dysregulation is involved in the etiology and progression of various human diseases. Formalin-fixed paraffin-embedded (FFPE) samples represent the gold standard for archiving pathology samples, and thus FFPE samples are a major resource of samples in clinical research. However, chromatin-based epigenetic assays in the clinical settings are limited to fresh or frozen samples, and are hampered by low chromatin yield in FFPE samples due to the lack of a reliable and efficient chromatin preparation method. Here, we introduce a new chromatin extraction method from FFPE tissues (Chrom-EX PE) for chromatin-based epigenetic assays. RESULTS: During rehydration of FFPE tissues, applying a tissue-level cross-link reversal into the deparaffinized tissue at 65 °C dramatically increased chromatin yield in the soluble fraction. The resulting chromatin is compatible with targeted ChIP-qPCR and genome-wide ChIP-seq approaches. The chromatin prepared by Chrom-EX PE showed a gradual fragmentation pattern with varying incubation temperature. At temperatures below 37 °C, the majority of soluble chromatin is over 1 kb. The soluble chromatin prepared in the range of 45-60 °C showed a typical nucleosomal pattern. And the majority of chromatin prepared at 65 °C is close to mononucleosomal size. These observations indicate that chromatin preparation from FFPE samples can be controlled for downstream chromatin-based epigenetic assays. CONCLUSIONS: This study provided a new method that achieves efficient extraction of high-quality chromatin suitable for chromatin-based epigenetic assays with less damage on chromatin. This approach may provide a way to circumvent the over-fixed nature of FFPE tissues for future technology development.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Cromatina/aislamiento & purificación , Adhesión en Parafina/métodos , Secuenciación Completa del Genoma/métodos , Animales , Cromatina/genética , Epigénesis Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Temperatura , Fijación del Tejido
19.
Gastroenterology ; 154(8): 2122-2136.e12, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29501441

RESUMEN

BACKGROUND & AIMS: Muscularis propria macrophages lie close to cells that regulate gastrointestinal motor function, including interstitial cells of Cajal (ICC) and myenteric neurons. In animal models of diabetic gastroparesis, development of delayed gastric emptying has been associated with loss of macrophages that express cytoprotective markers and reduced networks of ICC. Mice with long-term diabetes and normal gastric emptying have macrophages that express anti-inflammatory markers and have normal gastric ICC. Mice homozygous for the osteopetrosis spontaneous mutation in the colony-stimulating factor 1 gene (Csf1op/op) do not have macrophages; when they are given streptozotocin to induce diabetes, they do not develop delayed gastric emptying. We investigated whether population of the gastric muscularis propria of diabetic Csf1op/op mice with macrophages is necessary to change gastric emptying, ICC, and myenteric neurons and investigated the macrophage-derived factors that determine whether diabetic mice do or do not develop delayed gastric emptying. METHODS: Wild-type and Csf1op/op mice were given streptozotocin to induce diabetes. Some Csf1op/op mice were given daily intraperitoneal injections of CSF1 for 7 weeks; gastric tissues were collected and cellular distributions were analyzed by immunohistochemistry. CD45+, CD11b+, F4/80+ macrophages were dissociated from gastric muscularis propria, isolated by flow cytometry and analyzed by quantitative real-time polymerase chain reaction. Cultured gastric muscularis propria from Csf1op/op mice was exposed to medium that was conditioned by culture with bone marrow-derived macrophages from wild-type mice. RESULTS: Gastric muscularis propria from Csf1op/op mice given CSF1 contained macrophages; 11 of 15 diabetic mice given CSF1 developed delayed gastric emptying and had damaged ICC. In non-diabetic Csf1op/op mice, administration of CSF1 reduced numbers of gastric myenteric neurons but did not affect the proportion of nitrergic neurons or ICC. In diabetic Csf1op/op mice given CSF1 that developed delayed gastric emptying, the proportion of nitrergic neurons was the same as in non-diabetic wild-type controls. Medium conditioned by macrophages previously exposed to oxidative injury caused damage to ICC in cultured gastric muscularis propria from Csf1op/op mice; neutralizing antibodies against IL6R or TNF prevented this damage to ICC. CD45+, CD11b+, and F4/80+ macrophages isolated from diabetic wild-type mice with delayed gastric emptying expressed higher levels of messenger RNAs encoding inflammatory markers (IL6 and inducible nitric oxide synthase) and lower levels of messenger RNAs encoding markers of anti-inflammatory cells (heme oxygenase 1, arginase 1, and FIZZ1) than macrophages isolated from diabetic mice with normal gastric emptying. CONCLUSIONS: In studies of Csf1op/op and wild-type mice with diabetes, we found delayed gastric emptying to be associated with increased production of inflammatory factors, and reduced production of anti-inflammatory factors, by macrophages, leading to loss of ICC.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Vaciamiento Gástrico/fisiología , Gastroparesia/fisiopatología , Macrófagos/fisiología , Estómago/fisiopatología , Animales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Gastroparesia/etiología , Humanos , Inmunohistoquímica , Células Intersticiales de Cajal/fisiología , Factor Estimulante de Colonias de Macrófagos/genética , Ratones , Músculo Liso/citología , Músculo Liso/patología , Músculo Liso/fisiopatología , Mutación , Estómago/citología , Estómago/patología , Estreptozocina/toxicidad
20.
Drug Metab Dispos ; 47(9): 983-994, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31292129

RESUMEN

Greater than 90% of significant genome-wide association study (GWAS) single-nucleotide polymorphisms (SNPs) are in noncoding regions of the genome, but only 25.6% are known expression quantitative trait loci (eQTLs). Therefore, the function of many significant GWAS SNPs remains unclear. We have identified a novel type of eQTL for which SNPs distant from ligand-activated transcription factor (TF) binding sites can alter target gene expression in a SNP genotype-by-ligand-dependent fashion that we refer to as pharmacogenomic eQTLs (PGx-eQTLs)-loci that may have important pharmacotherapeutic implications. In the present study, we integrated chromatin immunoprecipitation-seq with RNA-seq and SNP genotype data for a panel of lymphoblastoid cell lines to identify 10 novel cis PGx-eQTLs dependent on the ligand-activated TF aryl hydrocarbon receptor (AHR)-a critical environmental sensor for xenobiotic (drug) and immune response. Those 10 cis PGx-eQTLs were eQTLs only after AHR ligand treatment, even though the SNPs did not create/destroy an AHR response element-the DNA sequence motif recognized and bound by AHR. Additional functional studies in multiple cell lines demonstrated that some cis PGx-eQTLs are functional in multiple cell types, whereas others displayed SNP-by-ligand-dependent effects in just one cell type. Furthermore, four of those cis PGx-eQTLs had previously been associated with clinical phenotypes, indicating that those loci might have the potential to inform clinical decisions. Therefore, SNPs across the genome that are distant from TF binding sites for ligand-activated TFs might function as PGx-eQTLs and, as a result, might have important clinical implications for interindividual variation in drug response. SIGNIFICANCE STATEMENT: More than 90% of single-nucleotide polymorphisms (SNPs) that are associated with clinical phenotypes are located in noncoding regions of the genome. However, the mechanisms of action of many of those SNPs have not been elucidated, and drugs may unmask functional expression quantitative trail loci (eQTLs). In the current study, we used drugs that bind to the ligand-activated transcription factor aryl hydrocarbon receptor (AHR) and identified SNPs that were associated with interindividual variation in gene expression following drug exposure-termed pharmacogenomic (PGx)-eQTLs. Possibly of greater significance, those PGx-eQTL SNPs were outside of AHR binding sites, indicating that they do not interrupt AHR DNA recognition. PGx-eQTLs such as those described in this work may have crucial implications for interindividual variation in drug.


Asunto(s)
Variación Biológica Poblacional , Genoma Humano/genética , Sitios de Carácter Cuantitativo , Receptores de Hidrocarburo de Aril/genética , Xenobióticos/farmacocinética , Sitios de Unión , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Ligandos , Polimorfismo de Nucleótido Simple , RNA-Seq , Receptores de Hidrocarburo de Aril/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA