Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Exp Clin Cancer Res ; 43(1): 90, 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38523299

RESUMEN

BACKGROUND: Ropivacaine, a local anesthetic, exhibits anti-tumor effects in various cancer types. However, its specific functions and the molecular mechanisms involved in breast cancer cell stemness remain elusive. METHODS: The effects of ropivacaine on breast cancer stemness were investigated by in vitro and in vivo assays (i.e., FACs, MTT assay, mammosphere formation assay, transwell assays, western blot, and xenograft model). RNA-seq, bioinformatics analysis, Western blot, Luciferase reporter assay, and CHIP assay were used to explore the mechanistic roles of ropivacaine subsequently. RESULTS: Our study showed that ropivacaine remarkably suppressed stem cells-like properties of breast cancer cells both in vitro and in vivo. RNA-seq analysis identified GGT1 as the downstream target gene responding to ropivacaine. High GGT1 levels are positively associated with a poor prognosis in breast cancer. Ropivacaine inhibited GGT1 expression by interacting with the catalytic domain of AKT1 directly to impair its kinase activity with resultant inactivation of NF-κB. Interestingly, NF-κB can bind to the promoter region of GGT1. KEGG and GSEA analysis indicated silence of GGT1 inhibited activation of NF-κB signaling pathway. Depletion of GGT1 diminished stem phenotypes of breast cancer cells, indicating the formation of NF-κB /AKT1/GGT1/NF-κB positive feedback loop in the regulation of ropivacaine-repressed stemness in breast cancer cells. CONCLUSION: Our finding revealed that local anesthetic ropivacaine attenuated breast cancer stemness through AKT1/GGT1/NF-κB signaling pathway, suggesting the potential clinical value of ropivacaine in breast cancer treatment.


Asunto(s)
Neoplasias de la Mama , FN-kappa B , Humanos , Femenino , FN-kappa B/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Ropivacaína/farmacología , Ropivacaína/uso terapéutico , Anestésicos Locales/farmacología , Anestésicos Locales/uso terapéutico , Línea Celular Tumoral , Proteínas Proto-Oncogénicas c-akt/metabolismo
2.
Front Oncol ; 12: 1057147, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36531063

RESUMEN

Previously we uncovered the epigenetic regulation of medulloblastoma that low levels of H3K27me3 are required for Shh target gene expression and medulloblastoma growth. Since Jmjd3, an H3K27me3 demethylase, is responsible for maintaining low H3K27me3 at Shh target genes, targeting Jmjd3 could be an efficient way to inhibit Shh signaling and medulloblastoma growth. Here we show that the small molecule GSK-J4, an inhibitor of Jmjd3, significantly inhibited the expression of Shh target genes in Shh responsive cell models and primary cerebellar granule neuron precursors. GSK-J4 also significantly reduced the growth of primary Shh medulloblastoma cultures. Treating human medulloblastoma cell line DaoY by GSK-J4 led to cell cycle arrest at G0/G1 phase with decreased cells in S-phase. Tumor cell proliferation was significantly inhibited by GSK-J4 treatment. Gene expression analyses showed that GSK-J4 additionally constrained the expression of key genes in cholesterol biosynthesis. Our results highlight the possibility that targeting H3K27me3 demethylase Jmjd3 with GSK-J4 to inhibit Shh signaling and cholesterol metabolism is a potential application to treat Shh medulloblastoma.

3.
Proc Natl Acad Sci U S A ; 119(32): e2208855119, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35914126

RESUMEN

Wild-type (WT) mice maintain viable levels of blood glucose even when adipose stores are depleted by 6 d of 60% calorie restriction followed by a 23-h fast (hereafter designated as "starved" mice). Survival depends on ghrelin, an octanoylated peptide hormone. Mice that lack ghrelin suffer lethal hypoglycemia when subjected to the same starvation regimen. Ghrelin is known to stimulate secretion of growth hormone (GH), which in turn stimulates secretion of IGF-1 (insulin-like growth factor-1). In the current study, we found that starved ghrelin-deficient mice had a 90% reduction in plasma IGF-1 when compared with starved WT mice. Injection of IGF-1 in starved ghrelin-deficient mice caused a twofold increase in glucose production and raised blood glucose to levels seen in starved WT mice. Increased glucose production was accompanied by increases in plasma glycerol, fatty acids and ketone bodies, and hepatic triglycerides. All of these increases were abolished when the mice were treated with atglistatin, an inhibitor of adipose tissue triglyceride lipase. We conclude that IGF-1 stimulates adipose tissue lipolysis in starved mice and that this lipolysis supplies energy and substrates that restore hepatic gluconeogenesis. This action of IGF-1 in starved mice is in contrast to its known action in inhibiting adipose tissue lipase in fed mice. Surprisingly, the ghrelin-dependent maintenance of plasma IGF-1 in starved mice was not mediated by GH. Direct injection of GH into starved ghrelin-deficient mice failed to increase plasma IGF-1. These data call attention to an unsuspected role of IGF-1 in the adaptation to starvation.


Asunto(s)
Glucemia , Factor I del Crecimiento Similar a la Insulina , Inanición , Adaptación Fisiológica , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/enzimología , Tejido Adiposo/metabolismo , Animales , Glucemia/metabolismo , Ácidos Grasos/sangre , Ghrelina/metabolismo , Gluconeogénesis , Glicerol/sangre , Hormona del Crecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/análisis , Factor I del Crecimiento Similar a la Insulina/metabolismo , Cuerpos Cetónicos/sangre , Lipasa/antagonistas & inhibidores , Lipasa/metabolismo , Lipólisis , Hígado/metabolismo , Ratones , Compuestos de Fenilurea/farmacología , Inanición/sangre , Inanición/metabolismo , Triglicéridos/metabolismo
4.
Mol Neurobiol ; 59(9): 5766-5784, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35796899

RESUMEN

Programmed cell death (PCD) is an important pathologic process after spinal cord injury (SCI). As a new type of PCD, ferroptosis is involved in the secondary SCI. However, the underlying molecular mechanism remains unclear. In this study, we validated ferroptotic phenotype in an animal model of SCI. Then, the bioinformatic analyses performed on a microarray data of SCI (GSE45006). KEGG analysis suggested that the pathways of mTOR, HIF-1, VEGF, and protein process in endoplasmic reticulum were involved in SCI-induced ferroptosis. GO analysis revealed that oxidative stress, amide metabolic process, cation transport, and cytokine production were essential biological processes in ferroptosis after SCI. We highlighted five genes including ATF-3, XBP-1, HMOX-1, DDIT-3, and CHAC-1 as ferroptotic key gene in SCI. These results contribute to exploring the ferroptotic mechanism underlying the secondary SCI and providing potential targets for clinical treatment.


Asunto(s)
Ferroptosis , Traumatismos de la Médula Espinal , Animales , Apoptosis , Biología Computacional , Estrés Oxidativo , Médula Espinal/patología , Traumatismos de la Médula Espinal/tratamiento farmacológico
5.
Cancer Res ; 82(16): 2874-2886, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35731926

RESUMEN

Intratumor epigenetic heterogeneity is emerging as a key mechanism underlying tumor evolution and drug resistance. Epigenetic abnormalities frequently occur in medulloblastoma, the most common childhood malignant brain tumor. Medulloblastoma is classified into four subtypes including SHH medulloblastoma, which is characterized by elevated sonic hedgehog (SHH) signaling and a cerebellum granule neuron precursor (CGNP) cell-of-origin. Here, we report that the histone H3K27 methyltransferase polycomb repressor complex 2 (PRC2) is often heterogeneous within individual SHH medulloblastoma tumors. In mouse models, complete deletion of the PRC2 core subunit EED inhibited medulloblastoma growth, while a mosaic deletion of EED significantly enhanced tumor growth. EED is intrinsically required for CGNP maintenance by inhibiting both neural differentiation and cell death. Complete deletion of EED led to CGNP depletion and reduced occurrence of medulloblastoma. Surprisingly, medulloblastomas with mosaic EED levels grew faster than control wild-type tumors and expressed increased levels of oncogenes such as Igf2, which is directly repressed by PRC2 and has been demonstrated to be both necessary and sufficient for SHH medulloblastoma progression. Insulin-like growth factor 2 (IGF2) mediated the oncogenic effects of PRC2 heterogeneity in tumor growth. Assessing clones of a human medulloblastoma cell line with different EED levels confirmed that EEDlow cells can stimulate the growth of EEDhigh cells through paracrine IGF2 signaling. Thus, PRC2 heterogeneity plays an oncogenic role in medulloblastoma through both intrinsic growth competence and non-cell autonomous mechanisms in distinct tumor subclones. SIGNIFICANCE: The identification of an oncogenic function of PRC2 heterogeneity in medulloblastoma provides insights into subclone competition and cooperation during heterogeneous tumor evolution.


Asunto(s)
Neoplasias Cerebelosas , Proteínas de Drosophila , Meduloblastoma , Animales , Neoplasias Cerebelosas/patología , Cerebelo , Niño , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Meduloblastoma/patología , Ratones , Proteínas del Grupo Polycomb/metabolismo , Transducción de Señal/fisiología
6.
Reprod Sci ; 29(2): 646-651, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34159570

RESUMEN

Acephalic spermatozoa syndrome (ASS) is a severe form of teratozoospermia, previous studies have shown that SUN5 mutations are the major cause of acephalic spermatozoa syndrome. This study is to identify the pathogenic mutations in SUN5 leading to ASS. PCR and Sanger sequence were performed to define the breakpoints and mutations in SUN5. Whole genome sequencing (WGS) was performed to detect heterozygous deletion. Western blotting and immunofluorescence analysis detected the expression level and localization of SUN5. Furthermore, the pathogenicity of the mutant SUN5 was predicted in silico and was verified by the experiments in vitro. We identified one novel homozygous missense mutation (c.775G>A; p.G259S) and one compound heterozygous including one reported missense mutation (c.1043A>T; p.N348I) and a large deletion that contains partial EFCAB8 ( NM_001143967 .1) and BPIFB2 ( NM_025227 ) and complete SUN5 ( NM_080675 ), and one recurrent homozygous splice-site mutation (c.340G>A; p.G114R) in SUN5 in three patients with ASS. Our results showed that SUN5 could not be detected in the patients' spermatozoa and the exogenous expression level of the mutant protein was decreased in transfected HEK-293T cells. This study expands the mutational spectrum of SUN5. We recommended a clinical diagnostic strategy for SUN5 genomic deletion to screen heterozygous deletions and indicated that the diagnostic value of screening for SUN5 mutations and deletions in infertile men with ASS.


Asunto(s)
Infertilidad Masculina/genética , Proteínas de la Membrana/genética , Teratozoospermia/genética , Adulto , Western Blotting , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Masculino , Mutación Missense , Linaje , Eliminación de Secuencia/genética , Espermatozoides/metabolismo , Síndrome , Secuenciación Completa del Genoma
7.
J Assist Reprod Genet ; 39(1): 261-266, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34854019

RESUMEN

PURPOSE: To identify the genetic causes of multiple morphological anomalies of the flagella (MMAF) and oligoasthenoteratozoospermia (OAT). METHODS: Whole-exome sequencing (WES) was performed on the proband to identify pathogenic mutation for infertility. Western blotting and immunofluorescence analysis detected the expression level and localization of adenylate kinase 7 (AK7). RESULTS: We identified a novel homozygous missense mutation (NM_152327: c.1846G > A; p.E616K) in AK7 in two brothers with MMAF and OAT from a consanguineous family by WES. Western blotting and immunofluorescence experiments determined that the expression level of AK7 decreased in the sperm from the proband. The proband and his wife underwent two cycles of intracytoplasmic sperm injection (ICSI) treatment but got unfavorable outcomes. CONCLUSION: This study could provide precise genetic diagnosis for the patient and expand the spectrum of AK7 mutations.


Asunto(s)
Adenilato Quinasa/genética , Flagelos/genética , Mutación Missense/genética , Oligospermia/etiología , Adenilato Quinasa/efectos adversos , Adulto , Flagelos/metabolismo , Flagelos/microbiología , Humanos , Masculino , Oligospermia/genética , Oligospermia/fisiopatología
8.
J Assist Reprod Genet ; 38(10): 2791-2799, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34409526

RESUMEN

PURPOSE: To identify the genetic causes for acephalic spermatozoa syndrome. METHODS: Whole-exome sequencing was performed on the proband from a non-consanguineous to identify pathogenic mutations for acephalic spermatozoa syndrome. Quantitative real-time polymerase chain reaction and whole genome sequencing were subjected to detect deletion. The functional effect of the identified splicing mutation was investigated by minigene assay. Western blot and immunofluorescence were performed to detect the expression level and localization of mutant TSGA10 protein. RESULTS: Here, we identified a novel heterozygous splicing mutation in TSGA10 (NM_025244: c.1108-1G > T), while we confirmed that there was a de novo large deletion in the proband. The splicing mutation led to the skipping of the exon15 of TSGA10, which resulted in a truncated protein (p. A370Efs*293). Therefore, we speculated that the splicing mutation might affect transcription and translation without the dosage compensation of a normal allele, which possesses a large deletion including intact TSGA10. Western blot and immunofluorescence demonstrated that the very low expression level of truncated TSGA10 protein led the proband to present the acephalic spermatozoa phenotype. CONCLUSION: Our finding expands the spectrum of pathogenic TSGA10 mutations that are responsible for ASS and male infertility. It is also important to remind us of paying attention to the compound heterozygous deletion in patients from non-consanguineous families, so that we can provide more precise genetic counseling for patients.


Asunto(s)
Proteínas del Citoesqueleto/genética , Eliminación de Gen , Infertilidad Masculina/patología , Mutación , Empalme del ARN , Espermatozoides/anomalías , Teratozoospermia/patología , Femenino , Humanos , Infertilidad Masculina/genética , Masculino , Linaje , Pronóstico , Teratozoospermia/genética
9.
Cell Rep ; 36(2): 109357, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260936

RESUMEN

Neuronal activity-induced enhancers drive gene activation. We demonstrate that BRG1, the core subunit of SWI/SNF-like BAF ATP-dependent chromatin remodeling complexes, regulates neuronal activity-induced enhancers. Upon stimulation, BRG1 is recruited to enhancers in an H3K27Ac-dependent manner. BRG1 regulates enhancer basal activities and inducibility by affecting cohesin binding, enhancer-promoter looping, RNA polymerase II recruitment, and enhancer RNA expression. We identify a serine phosphorylation site in BRG1 that is induced by neuronal stimulations and is sensitive to CaMKII inhibition. BRG1 phosphorylation affects its interaction with several transcription co-factors, including the NuRD repressor complex and cohesin, possibly modulating BRG1-mediated transcription outcomes. Using mice with knockin mutations, we show that non-phosphorylatable BRG1 fails to efficiently induce activity-dependent genes, whereas phosphomimic BRG1 increases enhancer activity and inducibility. These mutant mice display anxiety-like phenotypes and altered responses to stress. Therefore, we reveal a mechanism connecting neuronal signaling to enhancer activities through BRG1 phosphorylation.


Asunto(s)
ADN Helicasas/genética , ADN Helicasas/metabolismo , Elementos de Facilitación Genéticos/genética , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Acetilación , Secuencia de Aminoácidos , Animales , Ansiedad/patología , Conducta Animal , Encéfalo/patología , ADN Helicasas/química , Células HEK293 , Histonas/metabolismo , Humanos , Lisina/metabolismo , Ratones Endogámicos C57BL , Mutación/genética , Proteínas Nucleares/química , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Estrés Psicológico/complicaciones , Factores de Transcripción/química , alfa-Fetoproteínas/metabolismo
10.
Clin Genet ; 100(3): 324-328, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33966269

RESUMEN

The genetic causes in most of patients with oocyte maturation arrest remain largely unknown. In this study, we identified a homozygous missense mutation (c.895T>C; p.C299R) in TBPL2 (TATA box binding protein like 2) in two infertile sisters with oocyte maturation arrest and degeneration from a consanguineous family by whole-exome sequencing. The TBPL2 mutation is rare and pathogenic, and impaired the transcription initiation function of the protein. Our results showed that TBPL2 mutation might be associated with female infertility due to oocyte maturation arrest and degeneration.


Asunto(s)
Infertilidad Femenina/genética , Mutación Missense , Proteínas Nucleares/genética , Oogénesis/genética , Proteínas Similares a la Proteína de Unión a TATA-Box/genética , Adulto , Muerte Celular/genética , Consanguinidad , Femenino , Homocigoto , Humanos , Linaje , Secuenciación del Exoma
11.
Proteomics ; 21(7-8): e2000144, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33570763

RESUMEN

C-C motif chemokine 18 (CCL18) belongs to the chemokine CC family and is predominantly secreted by M2-tumor-associated macrophages. It has been reported to be associated with various diseases and malignancies. Previous studies showed that CCL18 promotes metastasis by activating downstream kinases. However, it remains unknown whether CCL18 regulates post-translational modifications, other than phosphorylation, during tumorigenesis. Here, we demonstrate that CCL18 is up-regulated in non-small cell lung cancer (NSCLC) and is involved in regulating the lysine acetylome in A549 cells. Using the combination of SILAC labeling and high-efficiency acetylation enrichment methods, we identified 1372 lysine acetylation (Kac) sites on 796 proteins in CCL18-treated A549 cells. Among the identified Kac sites, 147 from 126 proteins were down-regulated and seven from five proteins were up-regulated with fold changes more than two and the p-value less than 0.05. Bioinformatics analysis further showed that the proteins with down-regulated acetylation play critical roles in glycolysis, oxidative phosphorylation, tricarboxylic acid cycle, and pentose phosphate pathway in A549 cells. These results suggest that CCL18 may be involved in the development of NSCLC by regulating acetylation of the proteins in many fundamental cellular processes, especially the metabolic reprogramming of tumor cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Quimiocinas CC/fisiología , Neoplasias Pulmonares/metabolismo , Lisina/metabolismo , Procesamiento Proteico-Postraduccional , Células A549 , Acetilación , Humanos
12.
Cancer Lett ; 499: 188-200, 2021 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-33253789

RESUMEN

The deregulation of epigenetic pathways has been implicated as a critical step in tumorigenesis including in childhood brain tumor medulloblastoma. The H3K27me3 demethylase UTX/KDM6A plays important roles in development and is frequently mutated in various types of cancer. However, how UTX regulates tumor development remains largely unclear. Here, we report the generation of a UTX-deleted mouse model of SHH medulloblastoma that demonstrates the tumor suppressor functions of UTX, which could be antagonized by the deletion of another H3K27me3 demethylase JMJD3/KDM6B. Intriguingly, UTX deletion in cancerous cerebellar granule neuron precursors (CGNPs) resulted in the impaired recruitment of host CD8+ T cells to the tumor microenvironment through a non-cell autonomous mechanism. In both mouse medulloblastoma models and in human medulloblastoma cells, we showed that UTX activates Th1-type chemokines, which are responsible for T cell migration. Surprisingly, our results showed that the depletion of cytotoxic CD8+ T cells did not affect mouse medulloblastoma growth. Nevertheless, the UTX/chemokine/T cell recruitment pathway we identified may be applied to many other cancers and may be important for improving cancer immunotherapy. In addition, UTX is required for the expression of NeuroD2 in precancerous progenitors, which encodes a potent proneural transcription factor. Overexpression of NEUROD2 in CGNPs decreased cell proliferation and increased neuron differentiation. We showed that UTX deletion led to impaired neural differentiation, which could coordinate with active SHH signaling to accelerate medulloblastoma development. Thus, UTX regulates both cell-intrinsic oncogenic processes and the tumor microenvironment in medulloblastoma. Our study provides insights into both medulloblastoma development and context dependent functions of UTX in tumorigenesis.


Asunto(s)
Neoplasias Cerebelosas/genética , Histona Demetilasas/metabolismo , Meduloblastoma/genética , Proteínas Supresoras de Tumor/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Neoplasias Cerebelosas/inmunología , Neoplasias Cerebelosas/patología , Cerebelo/citología , Cerebelo/patología , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Técnicas de Silenciamiento del Gen , Histona Demetilasas/genética , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Meduloblastoma/inmunología , Meduloblastoma/patología , Ratones , Ratones Transgénicos , Células Madre Neoplásicas/patología , Neuronas/patología , Neuropéptidos/genética , Cultivo Primario de Células , Receptor Smoothened/genética , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Proteínas Supresoras de Tumor/genética
13.
Proc Natl Acad Sci U S A ; 116(15): 7449-7454, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30910968

RESUMEN

When mice are subjected to 60% calorie restriction for several days, they lose nearly all of their body fat. Although the animals lack energy stores, their livers produce enough glucose to maintain blood glucose at viable levels even after a 23-hour fast. This adaptation is mediated by a marked increase in plasma growth hormone (GH), which is elicited by an increase in plasma ghrelin, a GH secretagogue. In the absence of ghrelin, calorie-restricted mice develop hypoglycemia, owing to diminished glucose production. To determine the site of GH action, in the current study we used CRISPR/Cas9 and Cre recombinase technology to produce mice that lack GH receptors selectively in liver (L-Ghr-/- mice) or in adipose tissue (Fat-Ghr-/- mice). When subjected to calorie restriction and then fasted for 23 hours, the L-Ghr-/- mice, but not the Fat-Ghr-/- mice, developed hypoglycemia. The fall in blood glucose in L-Ghr-/- mice was correlated with a profound drop in hepatic triglycerides. Hypoglycemia was prevented by injection of lactate or octanoate, two sources of energy to support gluconeogenesis. Electron microscopy revealed extensive autophagy in livers of calorie-restricted control mice but not in L-Ghr-/- mice. We conclude that GH acts through its receptor in the liver to activate autophagy, preserve triglycerides, enhance gluconeogenesis, and prevent hypoglycemia in calorie-restricted mice, a model of famine.


Asunto(s)
Autofagia , Glucemia/metabolismo , Restricción Calórica , Hormona del Crecimiento/sangre , Hipoglucemia/sangre , Hígado/metabolismo , Inanición/sangre , Animales , Glucemia/genética , Enfermedad Crónica , Modelos Animales de Enfermedad , Hormona del Crecimiento/genética , Hipoglucemia/genética , Hígado/patología , Ratones , Ratones Noqueados , Inanición/genética , Inanición/patología
14.
PLoS Biol ; 15(8): e2002063, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28777795

RESUMEN

Hedgehog (Hh) signaling in vertebrates depends on primary cilia. Upon stimulation, Hh pathway components, including Gli transcription factors, accumulate at primary cilia to transduce the Hh signal, but the mechanisms underlying their ciliary targeting remains largely unknown. Here, we show that the PY-type nuclear localization signal (PY-NLS)/karyopherinß2 (Kapß2) nuclear import system regulates Gli ciliary localization and Hh pathway activation. Mutating the PY-NLS in Gli or knockdown of Kapß2 diminished Gli ciliary localization. Kapß2 is required for the formation of Gli activator (GliA) in wild-type but not in Sufu mutant cells. Knockdown of Kapß2 affected Hh signaling in zebrafish embryos, as well as in vitro cultured cerebellum granule neuron progenitors (CGNPs) and SmoM2-driven medulloblastoma cells. Furthermore, Kapß2 depletion impaired the growth of cultured medulloblastoma cells, which was rescued by Gli overexpression. Interestingly, Kapß2 is a transcriptional target of the Hh pathway, thus forming a positive feedback loop for Gli activation. Our study unravels the molecular mechanism and cellular machinery regulating Gli ciliary localization and identifies Kapß2 as a critical regulator of the Hh pathway and a potential drug target for Hh-driven cancers.


Asunto(s)
Cilios/metabolismo , Proteínas Hedgehog/metabolismo , Señales de Localización Nuclear , Proteína con Dedos de Zinc GLI1/metabolismo , beta Carioferinas/metabolismo , Animales , Animales Modificados Genéticamente , Neoplasias Cerebelosas/metabolismo , Embrión no Mamífero/metabolismo , Retroalimentación Fisiológica , Meduloblastoma/metabolismo , Ratones , Células 3T3 NIH , Pez Cebra
15.
Mol Cell Biol ; 36(1): 70-83, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26459759

RESUMEN

Synapse development requires normal neuronal activities and the precise expression of synapse-related genes. Dysregulation of synaptic genes results in neurological diseases such as autism spectrum disorders (ASD). Mutations in genes encoding chromatin-remodeling factor Brg1/SmarcA4 and its associated proteins are the genetic causes of several developmental diseases with neurological defects and autistic symptoms. Recent large-scale genomic studies predicted Brg1/SmarcA4 as one of the key nodes of the ASD gene network. We report that Brg1 deletion in early postnatal hippocampal neurons led to reduced dendritic spine density and maturation and impaired synapse activities. In developing mice, neuronal Brg1 deletion caused severe neurological defects. Gene expression analyses indicated that Brg1 regulates a significant number of genes known to be involved in synapse function and implicated in ASD. We found that Brg1 is required for dendritic spine/synapse elimination mediated by the ASD-associated transcription factor myocyte enhancer factor 2 (MEF2) and that Brg1 regulates the activity-induced expression of a specific subset of genes that overlap significantly with the targets of MEF2. Our analyses showed that Brg1 interacts with MEF2 and that MEF2 is required for Brg1 recruitment to target genes in response to neuron activation. Thus, Brg1 plays important roles in both synapse development/maturation and MEF2-mediated synapse remodeling. Our study reveals specific functions of the epigenetic regulator Brg1 in synapse development and provides insights into its role in neurological diseases such as ASD.


Asunto(s)
ADN Helicasas/metabolismo , Factores de Transcripción MEF2/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Sinapsis/metabolismo , Factores de Transcripción/metabolismo , Animales , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Cromatina/metabolismo , ADN Helicasas/genética , Espinas Dendríticas/genética , Hipocampo/metabolismo , Factores de Transcripción MEF2/genética , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Factores de Transcripción/genética
16.
Nat Commun ; 5: 5425, 2014 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-25370275

RESUMEN

The Sonic hedgehog (Shh) signalling pathway plays important roles during development and in cancer. Here we report a Shh-induced epigenetic switch that cooperates with Gli to control transcription outcomes. Before induction, poised Shh target genes are marked by a bivalent chromatin domain containing a repressive histone H3K27me3 mark and an active H3K4me3 mark. Shh activation induces a local switch of epigenetic cofactors from the H3K27 methyltransferase polycomb repressive complex 2 (PRC2) to an H3K27me3 demethylase Jmjd3/Kdm6b-centred coactivator complex. We also find that non-enzymatic activities of Jmjd3 are important and that Jmjd3 recruits the Set1/MLL H3K4 methyltransferase complexes in a Shh-dependent manner to resolve the bivalent domain. In vivo, changes of the bivalent domain accompanied Shh-activated cerebellar progenitor proliferation. Overall, our results reveal a regulatory mechanism that underlies the activation of Shh target genes and provides insight into the causes of various diseases and cancers exhibiting altered Shh signalling.


Asunto(s)
Epigénesis Genética , Proteínas Hedgehog/metabolismo , Meduloblastoma/genética , Animales , Células Cultivadas , Cerebelo/crecimiento & desarrollo , Femenino , N-Metiltransferasa de Histona-Lisina/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Meduloblastoma/metabolismo , Ratones Endogámicos C57BL , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Embarazo , Proteína con Dedos de Zinc GLI1
17.
Gene ; 515(2): 266-71, 2013 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-23262334

RESUMEN

The expression of the bovine acetyl-CoA carboxylase-alpha-encoding gene (ACACA) was known to be controlled by three promoters. Here, we characterized a fourth promoter (PI) located 41 kb upstream of the adjacent nutritionally-regulated promoter PIA on bovine chromosome 19. Our results showed that PI is an intergenic promoter driving expression of ACACA and conceivably, by analogy with the homologous genomic arrangement in sheep a component of the chromatin-modifying complex gene (TADA2L). 5'-RACE experiments defined the 3' boundary of the promoter and a novel exon 1 comprising 263 bp. It features at position +226 an ORF encoding an N-terminally extended ACC-α enzyme. The PI sequence is GC-rich, has no TATA box and CAAT box, similar to the homologous promoters in sheep, mouse and human. Expression profiles showed that PI is the promoter driving expression of the dominant ACACA-transcript in brain. Reporter gene assays in HC-11 cells indicated that deletion of extended promoter segments harboring putative cAMP response elements (CRE) clustered in the distal promoter region and specificity protein 1 (Sp1) attachment sites lowered PI activity.


Asunto(s)
Acetil-CoA Carboxilasa/genética , Encéfalo/enzimología , Regulación Enzimológica de la Expresión Génica , Regiones Promotoras Genéticas , Acetil-CoA Carboxilasa/metabolismo , Animales , Secuencia de Bases , Bovinos , Línea Celular , Femenino , Genes Reporteros , Luciferasas de Luciérnaga/biosíntesis , Luciferasas de Luciérnaga/genética , Ratones , Datos de Secuencia Molecular , Especificidad de Órganos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN
18.
BMC Mol Biol ; 13: 21, 2012 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-22738246

RESUMEN

BACKGROUND: The enzyme acetyl-CoA carboxylase-alpha (ACC-α) is rate limiting for de novo fatty acid synthesis. Among the four promoters expressing the bovine gene, promoter IA (PIA) is dominantly active in lipogenic tissues. This promoter is in principal repressed but activated under favorable nutritional conditions. Previous analyses already coarsely delineated the repressive elements on the distal promoter but did not resolve the molecular nature of the repressor. Knowledge about the molecular functioning of this repressor is fundamental to understanding the nutrition mediated regulation of PIA activity. We analyzed here the molecular mechanism calibrating PIA activity. RESULTS: We finely mapped the repressor binding sites in reporter gene assays and demonstrate together with Electrophoretic Mobility Shift Assays that nuclear factor-Y (NF-Y) and CCAAT/enhancer binding protein-ß (C/EBPß) each separately repress PIA activity by binding to their cognate low affinity sites, located on distal elements of the promoter. Simultaneous binding of both factors results in strongest repression. Paradoxically, over expression of NFY factors, but also - and even more so - of C/EBPß significantly activated the promoter when bound to high affinity sites on the proximal promoter. However, co-transfection experiments revealed that NF-Y may eventually diminish the strong stimulatory effect of C/EBPß at the proximal PIA in a dose dependent fashion. We validated by chromatin immunoprecipitation, that NF-Y and C/EBP factors may physically interact. CONCLUSION: The proximal promoter segment of PIA appears to be principally in an active state, since even minute concentrations of both, NF-Y and C/EBPß factors can saturate the high affinity activator sites. Higher factor concentrations will saturate the low affinity repressive sites on the distal promoter resulting in reduced and calibrated promoter activity. Based on measurements of the mRNA concentrations of those factors in different tissues we propose that the interplay of both factors may set tissue-specific limits for PIA activity.


Asunto(s)
Acetil-CoA Carboxilasa/metabolismo , Fenómenos Fisiológicos Nutricionales de los Animales/fisiología , Factor de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Bovinos/metabolismo , Ácidos Grasos/biosíntesis , Acetil-CoA Carboxilasa/genética , Animales , Bovinos/genética , Inmunoprecipitación de Cromatina , Mapeo Cromosómico , Ensayo de Cambio de Movilidad Electroforética , Regiones Promotoras Genéticas/genética
19.
Proc Natl Acad Sci U S A ; 108(31): 12758-63, 2011 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-21768360

RESUMEN

Sonic hedgehog (Shh) signaling plays diverse roles during animal development and adult tissue homeostasis through differential regulation of Gli family transcription factors. Dysregulated Shh signaling activities have been linked to birth defects and tumorigenesis. Here we report that Brg, an ATP-dependent chromatin remodeling factor, has dual functions in regulating Shh target gene expression. Using a Brg conditional deletion in Shh-responding neural progenitors and fibroblasts, we demonstrate that Brg is required both for repression of the basal expression and for the activation of signal-induced transcription of Shh target genes. In developing telencephalons deficient for Brg, Shh target genes were derepressed, whereas Brg-deleted cerebellar granule neuron precursors failed to respond to Shh to increase their proliferation. The repressor function of Brg was mediated through Gli3 and both the repressor and activator functions of Brg appeared to be independent of its ATPase activity. Furthermore, Brg facilitates Gli coactivator histone deacetylase (HDAC) binding to the regulatory regions of Shh target genes, providing a possible mechanism for its positive role in Shh signaling. Our results thus reveal that a complex chromatin regulation mechanism underlies the precise transcription outcomes of Shh signaling and its diverse roles during development.


Asunto(s)
ADN Helicasas/metabolismo , Proteínas Hedgehog/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , ADN Helicasas/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Proteínas Hedgehog/genética , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Immunoblotting , Hibridación in Situ , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Proteínas Nucleares/genética , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Telencéfalo/citología , Telencéfalo/embriología , Telencéfalo/metabolismo , Técnicas de Cultivo de Tejidos , Factores de Transcripción/genética , Proteína Gli3 con Dedos de Zinc
20.
Mol Immunol ; 48(6-7): 895-908, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21255844

RESUMEN

Pathogen contact induces quickly in Mammary Epithelial Cells (MEC) the expression of the proinflammatory cytokine IL-8 and delayed that of the bactericidal ß-defensin LAP. Both genes encoding these factors feature on their proximal promoter a composite NF-κB/CEBP binding site. We compare here in MEC the role of NF-κB and C/EBP factors in regulating basal and pathogen-induced expression of both genes from cattle. Abrogating NF-κB binding to that site by introduction of a single point mutation blocks promoter activity of both genes in reporter gene assays. Chromatin accessibility PCR and Chromatin immunoprecipitation reveal that the chromatin of the resting LAP promoter is tightly packed and NF-κB p50 homodimer binding prevails. Infection results in chromatin decompaction accompanied by predominant recruitment of NF-κB p65 for promoter activation. Overexpression of transcription factors confirms a stimulatory role of NF-κB p65 but also a repressive function of C/EBPß for LAP promoter activity. These factors reverse roles to control IL-8 expression. NF-κB p65 homodimers already reside on the resting IL-8 promoter and induction recruits NF-κB p50. Overexpression of both NF-κB factors represses the promoter in MEC, but not in HEK293 cells. Inhibitors of NF-κB activation and nuclear recruitment both tremendously increase basal and pathogen stimulated IL-8 mRNA concentrations in MEC. Mutation of the C/EBP-binding site blocks and overexpression of C/EBPß stimulates IL-8-promoter activity. Thus, the pathogen-induced fast activation of diverse transcription factors acting through a common promoter binding site is gene specifically differentiated into opposite functional significance for swiftly (IL-8) or slowly (LAP) induced genes in MEC.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Interleucina-8/genética , Glándulas Mamarias Animales/patología , Factor de Transcripción ReIA/metabolismo , beta-Defensinas/genética , Animales , Sitios de Unión , Bovinos , Cromatina , Células Epiteliales/microbiología , Escherichia coli/fisiología , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Femenino , Células HEK293 , Humanos , Cinética , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/microbiología , Ratones , Subunidad p50 de NF-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , beta-Defensinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA