Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 296
Filtrar
1.
Neurogastroenterol Motil ; 33(6): e14073, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33382180

RESUMEN

BACKGROUND: Calcitonin gene-related peptide (CGRP) is possibly involved in recruitment of mucosal mast cells (MCs) in the gut that may be associated with the development of irritable bowel syndrome (IBS), but the role of CGRP on the activation of MCs is still unknown. METHODS: Using RNA sequencing (RNA-seq), we examined differentially expressed genes (DEGs) in mouse MCs following CGRP treatment. The expression of key genes in colonic MCs and their relationship with CGRP-containing fibers were examined by immunofluorescence in chronic water-avoidance stress (WAS)-induced visceral hyperalgesia mice. KEY RESULTS: A total of 29 DEGs were found significantly changed with 28 upregulated and 1 downregulated following treatment of MCs with CGRP. Bioinformatics analysis showed that key higher DEGs included those associated with response to corticotropin-releasing hormone (CRH), regulation of transcription, MC activation, and proliferation. These processes are enriched for genes associated with stress-induced MC activation in IBS. Western blot verified changes in representative DEGs (Nr4a3, Crem, Gpr35, FosB, Sphlk1) and real-time cell analysis (RTCA) verified the MC proliferation. The vast majority of colonic MCs nearly CGRP-containing fibers in WAS mice overexpressed only Nr4a3 with little to no FosB, Gpr35, Sphlk1, or Crem expression. Nr4a3 knockdown may attenuate the promotion effect of CGRP on MC viability. CONCLUSIONS & INFERENCES: Our results suggest that CGRP is a critical regulator of key expressed genes in MC activation. Nr4a3 as a novel regulator of MC function may have an effect on stress-induced visceral hyperalgesia, and this may represent the novel target for drug development.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/biosíntesis , Colon/patología , Regulación de la Expresión Génica , Hiperalgesia/patología , Mastocitos/patología , Dolor Visceral/patología , Animales , Péptido Relacionado con Gen de Calcitonina/genética , Proliferación Celular , Biología Computacional , Hormona Liberadora de Corticotropina/metabolismo , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Femenino , Mucosa Intestinal/citología , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Receptores de Esteroides/biosíntesis , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/biosíntesis , Receptores de Hormona Tiroidea/genética , Estrés Psicológico
2.
Toxicology ; 439: 152477, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32360609

RESUMEN

We previously reported that exposure during gestation and lactation to a low dose of glyphosate-based herbicide (GBH) reduced the area and perimeter of male offspring mammary gland at postnatal day 60 (PND60), whereas a higher dose increased the longitudinal growth of the gland. Here, our aim was to assess whether perinatal exposure to GBH exhibits endocrine disruptive action in male mammary gland at an early time point (pre-puberty), which could be related to the changes observed after puberty. We also wanted to explore whether an early evaluation of the male rat mammary gland is appropriate to assess exposure to potential endocrine disrupting chemicals (EDCs). Pregnant rats were orally exposed, through the diet, to vehicle (saline solution), 3.5 or 350 mg/kg/day of GBH from gestational day 9 until weaning. At PND21, the male offspring were euthanized, and mammary gland samples were collected. The histology and proliferation index of the mammary glands were evaluated, and the mRNA expression of estrogen (ESR1) and androgen (AR) receptors, cyclin D1 (Ccnd1), amphiregulin (Areg), insulin-like growth factor 1 (IGF1), epidermal growth factor receptor (EGFR) and IGF1 receptor (IGF1R) were assessed. Moreover, the phosphorylated-Erk1/2 (p-ERK1/2) protein expression was determined. No differences were observed in mammary epithelial structures and AR expression between experimental groups; however, the proliferation index was reduced in GBH3.5-exposed males. This result was associated with decreased ESR1, Ccnd1, Areg, IGF1, EGFR and IGF1R mRNA expressions, as well as reduced p-Erk1/2 protein expression in these animals. ESR1, Ccnd1, IGF1R and EGFR expressions were also reduced in GBH350-exposed males. In conclusion, the mammary gland development of pre-pubertal male rats is affected by perinatal exposure to GBH. Although further studies are still needed to understand the molecular mechanisms involved in GBH350 exposure, the present results may explain the alterations observed in mammary gland growth of post-pubertal males exposed to low doses of GBH. Our results also suggest that early evaluation of the male rat mammary gland is useful in assessing exposure to potential EDCs. However, analysis of EDCs effects at later time points should not be excluded.


Asunto(s)
Disruptores Endocrinos/toxicidad , Glicina/análogos & derivados , Herbicidas/toxicidad , Glándulas Mamarias Animales/crecimiento & desarrollo , Actinas/metabolismo , Animales , Femenino , Glicina/toxicidad , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Glándulas Mamarias Animales/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Ratas , Ratas Wistar , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/genética , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Esteroides/biosíntesis , Glifosato
3.
Med Oncol ; 36(8): 66, 2019 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-31183633

RESUMEN

Nuclear receptor subfamily 4, group A, member 3 (NR4A3) is a member of the NR4A subgroup of orphan nuclear receptors, implicated in the regulation of diverse biological functions, including metabolism, angiogenesis, inflammation, cell proliferation, and apoptosis. Although many reports have suggested the involvement of NR4A3 in the development and/or progression of tumors, its role varies among tumor types. Previously, we reported that DNA hypomethylation at NR4A3 exon 3 is associated with lower survival rate of neuroblastoma (NB) patients. As hypomethylation of this region results in reduced expression of NR4A3, our observations suggested that NR4A3 functions as a tumor suppressor in NB. However, the exact mechanisms underlying its functions have not been clarified. In the present study, we analyzed public databases and showed that reduced NR4A3 expression was associated with shorter survival period of NB in two out of three datasets. An in vitro study revealed that forced expression of NR4A3 in human NB-derived cell line NB1 resulted in elongation of neurites along with overexpression of GAP43, one of the differentiation markers of NB. On the other hand, siRNA-mediated knockdown of NR4A3 suppressed the expression level of GAP43. Interestingly, the forced expression of NR4A3 induced only the GAP43 but not the other molecules involved in NB cell differentiation, such as MYCN, TRKA, and PHOX2B. These results indicated that NR4A3 directly activates the expression of GAP43 and induces differentiated phenotypes of NB cells, without affecting the upstream signals regulating GAP43 expression and NB differentiation.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Neuroblastoma/metabolismo , Receptores de Esteroides/biosíntesis , Receptores de Hormona Tiroidea/biosíntesis , Diferenciación Celular/fisiología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Proteína GAP-43/biosíntesis , Técnicas de Silenciamiento del Gen , Humanos , Neuritas/metabolismo , Neuritas/patología , Neuroblastoma/genética , Neuroblastoma/patología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética , Regulación hacia Arriba
4.
Am J Surg Pathol ; 43(9): 1264-1272, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31094928

RESUMEN

Recently, we discovered the recurrent genomic rearrangement [t(4;9)(q13;q31)] enabling upregulation of the transcription factor Nuclear Receptor Subfamily 4 Group A Member 3 (NR4A3) through enhancer hijacking as the oncogenic driver event in acinic cell carcinoma (AciCC) of the salivary glands. In the current study, we evaluated the usefulness of NR4A3 immunostaining and NR4A3 fluorescence in situ hybridization (FISH) in the differential diagnosis of AciCC, comparing a total of 64 AciCCs including 17% cases with high-grade transformation, 29 secretory (mammary analog) carcinomas (MASC), and 70 other salivary gland carcinomas. Nuclear NR4A3 immunostaining was a highly specific (100%) and sensitive (98%) marker for AciCC with only 1 negative case, whereas NR4A3 FISH was less sensitive (84%). None of the MASCs or other salivary gland carcinomas displayed any nuclear NR4A3 immunostaining. The recently described HTN3-MSANTD3 gene fusion was observed in 4 of 49 (8%) evaluable AciCCs, all with nuclear NR4A3 immunostaining. In summary, NR4A3 immunostaining is a highly specific and sensitive marker for AciCC, which may be especially valuable in cases with high-grade transformation and in "zymogen granule"-poor examples within the differential diagnostic spectrum of AciCC and MASC.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma de Células Acinares/diagnóstico , Proteínas de Unión al ADN/biosíntesis , Receptores de Esteroides/biosíntesis , Receptores de Hormona Tiroidea/biosíntesis , Neoplasias de las Glándulas Salivales/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Proteínas de Unión al ADN/análisis , Diagnóstico Diferencial , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Receptores de Esteroides/análisis , Receptores de Hormona Tiroidea/análisis , Sensibilidad y Especificidad , Adulto Joven
5.
Sci Rep ; 9(1): 4203, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862839

RESUMEN

The smaller tea tortrix, Adoxophyes honmai, has developed strong resistance to tebufenozide, a diacylhydrazine-type (DAH) insecticide. Here, we investigated its mechanism by identifying genes responsible for the tebufenozide resistance using various next generation sequencing techniques. First, double-digest restriction site-associated DNA sequencing (ddRAD-seq) identified two candidate loci. Then, synteny analyses using A. honmai draft genome sequences revealed that one locus contained the ecdysone receptor gene (EcR) and the other multiple CYP9A subfamily P450 genes. RNA-seq and direct sequencing of EcR cDNAs found a single nucleotide polymorphism (SNP), which was tightly linked to tebufenozide resistance and generated an amino acid substitution in the ligand-binding domain. The binding affinity to tebufenozide was about 4 times lower in in vitro translated EcR of the resistant strain than in the susceptible strain. RNA-seq analyses identified commonly up-regulated genes in resistant strains, including CYP9A and choline/carboxylesterase (CCE) genes. RT-qPCR analysis and bioassays showed that the expression levels of several CYP9A and CCE genes were moderately correlated with tebufenozide resistance. Collectively, these results suggest that the reduced binding affinity of EcR is the main factor and the enhanced detoxification activity by some CYP9As and CCEs plays a supplementary role in tebufenozide resistance in A. honmai.


Asunto(s)
Sistema Enzimático del Citocromo P-450 , Resistencia a Medicamentos , Hidrazinas/farmacología , Proteínas de Insectos , Insecticidas/farmacología , Lepidópteros , Receptores de Esteroides , Animales , Sistema Enzimático del Citocromo P-450/biosíntesis , Sistema Enzimático del Citocromo P-450/genética , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/genética , Regulación de la Expresión Génica/efectos de los fármacos , Estudio de Asociación del Genoma Completo , Proteínas de Insectos/biosíntesis , Proteínas de Insectos/genética , Lepidópteros/genética , Lepidópteros/metabolismo , Receptores de Esteroides/biosíntesis , Receptores de Esteroides/genética
6.
J Biol Chem ; 294(13): 4889-4897, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30696767

RESUMEN

The Nr4a subfamily of nuclear receptor comprises three members in mammalian cells: Nur77/Nr4a1, Nurr1/Nr4a2, and Nor1/Nr4a3. Nr4a proteins play key roles in the regulation of glucose homeostasis in peripheral metabolic tissues. However, their biological functions in ß-cells remain relatively uncharacterized. Here we sought to investigate the potential role of Nor1 in the regulation of ß-cell mass and, in particular, ß-cell survival/apoptosis. We used histological analysis to examine the consequences of genetic deletion of either Nur77 and Nor1 on ß-cell mass, investigated the expression patterns of Nr4as in human islets and INS cells and performed gain- and loss-of-function experiments to further characterize the role of Nor1 in ß-cell apoptosis. Surprisingly, Nor1 knockout mice displayed increased ß-cell mass, whereas mice with genetic deletion of Nur77 did not exhibit any significant differences compared with their WT littermates. The increase in ß-cell mass in Nor1 knockout mice was accompanied by improved glucose tolerance. A gene expression study performed in both human islets and INS cells revealed that Nor1 expression is significantly increased by pro-inflammatory cytokines and, to a lesser extent, by elevated concentrations of glucose. Nor1 overexpression in both INS and human islet cells caused apoptosis, whereas siRNA-mediated Nor1 knockdown prevented cytokine-induced ß-cell death. Finally, Nor1 expression was up-regulated in islets of individuals with type 2 diabetes. Altogether, our results uncover that Nor1 negatively regulates ß-cell mass. Nor1 represents a promising molecular target in diabetes treatment to prevent ß-cell destruction.


Asunto(s)
Apoptosis , Proteínas de Unión al ADN/biosíntesis , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Receptores de Esteroides/biosíntesis , Receptores de Hormona Tiroidea/biosíntesis , Regulación hacia Arriba , Animales , Citocinas , Proteínas de Unión al ADN/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Humanos , Células Secretoras de Insulina/patología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética
7.
Toxicol Appl Pharmacol ; 359: 1-11, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30196065

RESUMEN

Environmental factors, particularly xenoestrogens adversely affect reproductive health and their main mechanism is based on steroid-signaling pathway alterations. The presence of bisphenol A (BPA) in the environment has been confirmed and it is about to be replaced by analogues such as bisphenol F (BPF) and bisphenol S (BPS). Whether the BPF and BPS exert similar adverse effects to BPA has become the subject of intense scientific scrutiny. The aim of the present study was to evaluate and compare the cellular, transcriptomic and methylome effects of exposure to BPA, BPF, BPS individually and in combination on GC-2 spermatocyte cell line. The results show that all studied compounds affect cell viability, induce apoptosis and cause cellular damage. BPA, BPF and BPS also influence GC-2 cell steroid receptor and steroidogenesis related genes expressions. In addition to specific molecular mechanisms, all studied compounds also increase global DNA methylation. Exposure to a combination of all the studied compounds caused comparable effects on cell culture to each of them examined separately. These data suggest that exposure to BPA and its main substitutes- BPF and BPS induced multitude of effects and hence, BPF and BPS are not safe alternative to BPA in terms of male reproductive health.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Disruptores Endocrinos/toxicidad , Fenoles/toxicidad , Espermatocitos/efectos de los fármacos , Sulfonas/toxicidad , Transcriptoma/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Receptores de Esteroides/biosíntesis , Receptores de Esteroides/efectos de los fármacos , Receptores de Esteroides/genética , Esteroides/biosíntesis
8.
J Cutan Pathol ; 45(12): 886-890, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30178542

RESUMEN

BACKGROUND: Primary cutaneous mucinous carcinoma (PCMC) is a rare epithelial tumor with unclear histogenesis. METHODS: We evaluated the immunohistochemical expression of the estrogen receptor (ER), progesterone receptor (PR), and androgen receptor (AR) in six cases of PCMC. The immunoreactivity of adipophilin and gross cystic disease fluid protein (GCDFP)-15 was investigated to determine the origin of the tumor. RESULTS: The study included five males and one female aged 50 to 69 years who presented with a cutaneous mass in the face. Immunoreactivity for ER, PR, and AR was observed in all cases, and all cases were negative for adipophilin but positive for GCDFP-15. CONCLUSIONS: This report is the first to show AR expression in PCMC. All of followed cases manifested indolent clinical course, and the prognostic significance of hormone receptors in PCMC remains unclear. The negative immunoreactivity of PCMC for adipophilin and positivity for GCDFP-15 suggests a more likely relationship to apocrine than to sebaceous glands.


Asunto(s)
Adenocarcinoma Mucinoso/metabolismo , Proteínas Portadoras/biosíntesis , Neoplasias Faciales/metabolismo , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/biosíntesis , Proteínas de Neoplasias/biosíntesis , Perilipina-2/biosíntesis , Receptores de Esteroides/biosíntesis , Neoplasias Cutáneas/metabolismo , Adenocarcinoma Mucinoso/patología , Anciano , Neoplasias Faciales/patología , Femenino , Humanos , Masculino , Proteínas de Transporte de Membrana , Persona de Mediana Edad , Neoplasias Cutáneas/patología
9.
World J Surg Oncol ; 16(1): 147, 2018 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-30025533

RESUMEN

BACKGROUND: In here, we evaluated pattern of metastasis and cross-compared clinicopathological features between different age groups with breast cancer (BC). METHODS: This study was conducted in the Shiraz Breast Cancer Registry (largest BC registry in Iran). Patients were classified as < 30 years old (group 1), 30-60 years old (group 2), and > 60 years old (group 3). The three age groups were compared regarding clinical and baseline characteristics. RESULTS: Overall, 564 individuals entered group 1, 4519 group 2, and 670 group 3. Group 1 had lower rates of tumor necrosis (p < 0.001), higher lymphatic or vascular invasion (p = 0.002), estrogen receptor-negative individuals, and HER2-positive individuals (p ≤ 0.001). Younger groups had more stage 3 BC (31.1, 25.6, and 19.7% for groups 1, 2, and 3, respectively) (p = 0.016), grade 3 BC (27.4, 20.6, and 16.5% for groups 1, 2, and 3, respectively) (p = 0.001), and grade 3 nucleus (43.1, 34.5, and 27.6% for groups 1, 2, and 3, respectively) (p < 0.001). Group 1 had higher rates of regional metastasis (4.7 vs. 1.5 and 2.1% for groups 2 and 3, respectively). Younger individuals had higher rates of brain metastasis (13.3, 5.4, and 1.1% for groups 1, 2, and 3, respectively). Moreover, those > 60 years old had more lung metastasis (33 vs. 12.6 and 6.7% for groups 2 and 1, respectively) (p < 0.001). Younger groups had more < 5-year recurrence (16.3, 11.7, and 8.9%, for groups 1, 2, and 3, respectively) (p = 0.023). CONCLUSION: Pattern and site of recurrence changes according to age in BC. This brings up the question whether age is an independent predictor of organ of metastasis or is site of metastasis the result of other clinicopathological determinants which differ between age groups.


Asunto(s)
Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/secundario , Adulto , Factores de Edad , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/terapia , Estudios Transversales , Femenino , Humanos , Inmunohistoquímica , Irán/epidemiología , Metástasis Linfática , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Metástasis de la Neoplasia , Pronóstico , Receptor ErbB-2/biosíntesis , Receptores de Esteroides/biosíntesis , Sistema de Registros
10.
Mol Cell Biochem ; 448(1-2): 251-263, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29446045

RESUMEN

Dietary administration of orotic acid (OA), an intermediate in the pyrimidine biosynthetic pathway, is considered to provide a wide range of beneficial effects, including cardioprotection and exercise adaptation. Its mechanisms of action, when applied extracellularly, however, are barely understood. In this study, we evaluated potential effects of OA on skeletal muscle using an in vitro contraction model of electrically pulse-stimulated (EPS) C2C12 myotubes. By analyzing a subset of genes representing inflammatory, metabolic, and structural adaptation pathways, we could show that OA supplementation diminishes the EPS-provoked expression of inflammatory transcripts (interleukin 6, Il6; chemokine (C-X-C Motif) ligand 5, Cxcl5), and attenuated transcript levels of nuclear receptor subfamily 4 group A member 3 (Nr4A3), early growth response 1 (Egr1), activating transcription factor 3 (Atf3), and fast-oxidative MyHC-IIA isoform (Myh2). By contrast, OA had no suppressive effect on the pathogen-provoked inflammatory gene response in skeletal muscle cells, as demonstrated by stimulation of C2C12 myotubes with bacterial LPS. In addition, we observed a suppressive effect of OA on EPS-induced phosphorylation of AMP-activated protein kinase (AMPK), whereas EPS-triggered phosphorylation/activation of the mammalian target of rapamycin (mTOR) was not affected. Finally, we demonstrate that OA positively influences glycogen levels in EP-stimulated myotubes. Taken together, our results suggest that in skeletal muscle cells, OA modulates both the inflammatory and the metabolic reaction provoked by acute contraction. These results might have important clinical implications, specifically in cardiovascular and exercise medicine.


Asunto(s)
Contracción Muscular/efectos de los fármacos , Mioblastos Esqueléticos/metabolismo , Ácido Orótico/farmacología , Factor de Transcripción Activador 3/biosíntesis , Animales , Quimiocina CXCL5/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Estimulación Eléctrica , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-6/biosíntesis , Ratones , Mioblastos Esqueléticos/citología , Proteínas del Tejido Nervioso/biosíntesis , Receptores de Esteroides/biosíntesis , Receptores de Hormona Tiroidea/biosíntesis , Serina-Treonina Quinasas TOR/biosíntesis
11.
PLoS One ; 13(2): e0193242, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29470550

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) is the sixth most frequent cancer worldwide. The pregnane X receptor (PXR) is a nuclear receptor regulating several target genes associated with cancer malignancy. We here demonstrated a significant effect of PXR on HNSCC cell growth, as evidenced in PXR knock-down experiments. PXR transcriptional activity is more importantly regulated by the presence of coactivators and corepressors than by PXR protein expression. To date, there is scarce information on the regulation of PXR in HNSCC and on its role in the pathogenesis of this disease. Coactivator and corepressor expression was screened through qRT-PCR in 8 HNSCC cell lines and correlated to PXR activity, determined by using a reporter gene assay. All cell lines considerably expressed all the cofactors assessed. PXR activity negatively correlated with nuclear receptor corepressor 2 (NCoR2) expression, indicating a major role of this corepressor in PXR modulation and suggesting its potential as a surrogate for PXR activity in HNSCC. To test the association of NCoR2 with the malignant phenotype, a subset of three cell lines was transfected with an over-expression plasmid for this corepressor. Subsequently, cell growth and chemoresistance assays were performed. To elucidate the mechanisms underlying NCoR2 effects on cell growth, caspase 3/7 activity and protein levels of cleaved caspase 3 and PARP were evaluated. In HNO97 cells, NCoR2 over-expression decreased cell growth, chemoresistance and increased cleaved caspase 3 levels, caspase activity and cleaved PARP levels. On the contrary, in HNO124 and HNO210 cells, NCoR2 over-expression increased cell growth, drug resistance and decreased cleaved caspase 3 levels, caspase activity and cleaved PARP levels. In conclusion, we demonstrated a role of PXR and NCoR2 in the modulation of cell growth in HNSCC. This may contribute to a better understanding of the highly variable HNSCC therapeutic response.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/metabolismo , Proteínas de Neoplasias/biosíntesis , Co-Represor 2 de Receptor Nuclear/biosíntesis , Receptores de Esteroides/biosíntesis , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 7/genética , Caspasa 7/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Proteínas de Neoplasias/genética , Co-Represor 2 de Receptor Nuclear/genética , Receptor X de Pregnano , Receptores de Esteroides/genética
12.
J Cell Biochem ; 119(8): 6514-6526, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29363789

RESUMEN

The somatic component of seminiferous epithelium, the Sertoli cells (Sc) respond to Follicle stimulating hormone (FSH), and Testosterone (T) to produce factors which are necessary for germ cell (Gc) survival and differentiation. Infant Sc do not support spermatogenesis in spite of sufficient hormonal milieu, a situation similar to that found in male idiopathic infertility. Sc maturation during pubertal period involves expression of some genes which may be important for initiation of spermatogenesis. Analysis of differentially expressed genes, one by one, in infant and pubertal Sc might provide useful information about the regulation of spermatogenesis. DNA microarray based analysis of mRNA from 5-days (infant) and 12-days (pubertal) old rat Sc revealed increased expression of Nor-1 by pubertal Sc. NOR-1 is an orphan nuclear receptor involved in maintaining cellular homeostasis and disease. We generated transgenic mice using shRNA cloned under Pem (Rhox5) promoter which is activated at puberty in Sc. Such transgenic mice had reduced Nor-1 expression and increased Tgfß1, Tgfß3, and Smad3 expression. Moreover, an increase in ß-catenin expression was observed in NOR-1 knockdown testes. High ß-catenin in such transgenic mice was found to be associated with disruption of Sc maturation characterized by elevated expression of Anti Mullerian hormone, Cytokeratin 18, and Sox9. This disruption of Sc maturation resulted in Gc apoptosis. Such NOR-1 knockdown mice showed reduced sperm count and litter size. We report for the first time that NOR-1 plays a crucial role in regulating sperm count and male fertility.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Fertilidad , Proteínas del Tejido Nervioso/biosíntesis , Receptores de Esteroides/biosíntesis , Receptores de Hormona Tiroidea/biosíntesis , Células de Sertoli/metabolismo , Espermatogénesis , Espermatozoides/metabolismo , Animales , Proteínas de Unión al ADN/genética , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Ratas , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética , Células de Sertoli/citología , Recuento de Espermatozoides , Espermatozoides/citología
13.
Mol Pharmacol ; 92(1): 48-56, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28408657

RESUMEN

Small noncoding microRNAs act as post-transcriptional regulators of gene expression involved in diverse biologic functions. Pregnane X receptor (PXR, NR1I2), a member of the superfamily of nuclear receptors, is a transcription factor governing the transport and biotransformation of various drugs and other chemicals. In the present study, we identified a specific microRNA (miR) involved in regulating the expression and functionality of human PXR (hPXR). According to bioinformatics analysis employing three commonly used algorithms (TargetScan, miRanda, and DIANA-microT-CDS), miR-18a-5p was predicted to be the top candidate microRNA regulator of hPXR. Consequently, this microRNA was selected for detailed experimental investigation. As shown in cell-based dual-luciferase reporter gene assays, functional interaction occurred between miR-18a-5p and the microRNA recognition element of miR-18a-5p in the 3'-untranslated region of hPXR mRNA. Transfection of LS180 human colorectal adenocarcinoma cells with an miR-18a-5p mimic decreased hPXR mRNA and protein expression, whereas transfection of LS180 cells with an miR-18a-5p inhibitor increased hPXR mRNA and protein expression. The decrease in hPXR expression by the miR-18a-5p mimic was associated with a reduction in the extent of hPXR target gene (CYP3A4) induction by rifampin and rilpivirine. Treatment of untransfected LS180 cells with either of these hPXR agonists decreased endogenous expression of miR-18a-5p, and this preceded the onset of CYP3A4 induction. In conclusion, miR-18a-5p is a negative regulator of hPXR expression and the hPXR agonists rifampin and rilpivirine are chemical suppressors of miR-18a-5p expression.


Asunto(s)
MicroARNs/antagonistas & inhibidores , MicroARNs/biosíntesis , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/biosíntesis , Rifampin/farmacología , Rilpivirina/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Expresión Génica , Humanos , Luciferasas de Renilla , MicroARNs/genética , Receptor X de Pregnano , Receptores de Esteroides/agonistas , Inhibidores de la Transcriptasa Inversa/farmacología
14.
Cancer Res ; 77(9): 2375-2386, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28249906

RESUMEN

Nuclear orphan receptor NR4A1 exerts an essential tumor suppressor function in aggressive lymphomas. In this study, we investigated the hypothesized contribution of the related NR4A family member NR4A3 to lymphomagenesis. In aggressive lymphoma patients, low expression of NR4A3 was associated with poor survival. Ectopic expression or pharmacological activation of NR4A3 in lymphoma cell lines led to a significantly higher proportion of apoptotic cells. In a mouse NSG xenograft model of lymphoma (stably transduced SuDHL4 cells), NR4A3 expression abrogated tumor growth, compared with vector control and uninduced cells that formed massive tumors. Transcript analysis of four different aggressive lymphoma cell lines overexpressing either NR4A3 or NR4A1 revealed that apoptosis was driven similarly by induction of BAK, Puma, BIK, BIM, BID, and Trail. Overall, our results showed that NR4A3 possesses robust tumor suppressor functions of similar impact to NR4A1 in aggressive lymphomas. Cancer Res; 77(9); 2375-86. ©2017 AACR.


Asunto(s)
Carcinogénesis/genética , Proliferación Celular/genética , Proteínas de Unión al ADN/genética , Linfoma/genética , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/biosíntesis , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Linfoma/patología , Masculino , Ratones , Receptores de Esteroides/biosíntesis , Receptores de Hormona Tiroidea/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Neurochem Res ; 42(8): 2167-2177, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28303499

RESUMEN

Drug-resistance epilepsy (DRE) is attributed to the brain P-glycoprotein (P-gp) overexpression. We previously reported that nuclear factor-kappa B (NF-κB) played a critical role in regulating P-gp expression at the brain of the acute seizure rats. This study was extended further to investigate the interaction effect of NF-κB and pregnane X receptor (PXR) on P-gp expression at the brain of chronic epileptic rats treated with carbamazepine (CBZ). The chronic epileptic models were induced by the micro-injection of kainic acid (KA) into rats' hippocampus. Subsequently, the successful models were treated with different intervention agents of CBZ; PMA(a non-specific PXR activity inhibitor) or PDTC(a specific NF-κB activity inhibitor) respectively. The expression levels of P-gp and its encoded gene mdr1a/b were significantly up-regulated on the brain of KA-induced chronic epilepsy rats or the epilepsy rats treated with CBZ for 1 week, meanwhile with a high expression of PXR. The treatment of PMA dramatically reduced both PXR and P-gp expressions at the protein and mRNA levels in the chronic epilepsy brain. By compared to the epilepsy model group, the P-gp expression was not markedly attenuated by the inhibition of NF-κB activity with PDTC treatment, nevertheless with a decrease of NF-κB expression in this intervention group. Higher levels of proinflammatory cytokines(IL-1ß, IL-6, TNF-α) were found both in the brain tissue and the serum in the epilepsy rats of each group. There was a declined trend of the pro-inflammatory cytokines expression of the PDTC treatment group but with no statistical significance. This study demonstrates for the first time that P-gp up-regulation is due to increase PXR expression in the chronic phase of epilepsy, differently from that NF-κB signaling may induce the P-gp expression in the acute seizure phase. Our results offer insights into the mechanism underlying the development of DRE using or not using CBZ treatment.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Encéfalo/metabolismo , Epilepsia/metabolismo , Ácido Kaínico/toxicidad , FN-kappa B/biosíntesis , Receptores de Esteroides/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Encéfalo/efectos de los fármacos , Epilepsia/inducido químicamente , Epilepsia/genética , Expresión Génica , Masculino , Receptor X de Pregnano , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
16.
Br J Pharmacol ; 174(8): 700-717, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28128444

RESUMEN

BACKGROUND AND PURPOSE: Imatinib mesylate (IM) is a first-line treatment for chronic myeloid leukaemia (CML) as a specific inhibitor of BCR-ABL tyrosine kinase. As IM is widely used in CML, in combination with other drugs, the effects of IM on drug-metabolizing enzymes (DMEs) are crucial to the design of rational drug administration. Carboxylesterases (CESs) are enzymes catalysing the hydrolytic biotransformation of several clinically useful drugs. Although IM is known to inhibit cytochromes P450 (CYPs), its effects on DMEs, and CESs in particular, are still largely undefined. EXPERIMENTAL APPROACH: Hepatoma cell lines (HepG2 and Huh7) and primary mouse hepatocytes were used. mRNA and protein expression were evaluated by quantitative RT-PCR and Western blot analysis. Reporter luciferase activity was determined by transient co-transfection experiment. Pregnane X receptor (PXR) expression was regulated by overexpression and RNA interference. The activity of CESs was determined by enzymic and toxicological assays. Mice were treated with a range of doses of IM to analyse expression of CESs in mouse liver. KEY RESULTS: The expression and activity of CESs were markedly repressed by IM, along with the down-regulation of PXR and inhibited expression and activity of CYP3A4 and P-gp. CONCLUSIONS AND IMPLICATIONS: Down-regulation of PXR mediates IM-induced suppression of CESs. IM may inhibit expression of other genes targeted by PXR, thus inducing a wide range of potential drug-drug interactions during treatment of CML. The data deserve further elucidation including clinical trials.


Asunto(s)
Antineoplásicos/farmacología , Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Regulación hacia Abajo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Mesilato de Imatinib/farmacología , Receptores de Esteroides/biosíntesis , Receptores de Esteroides/deficiencia , Animales , Antineoplásicos/administración & dosificación , Hidrolasas de Éster Carboxílico/metabolismo , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Humanos , Mesilato de Imatinib/administración & dosificación , Masculino , Ratones , Ratones Endogámicos ICR , Receptor X de Pregnano , Receptores de Esteroides/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
17.
Int J Mol Sci ; 17(10)2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27775588

RESUMEN

BACKGROUND: There is an abnormal expression of nuclear receptor subfamily 2 group F member 6 (NR2F6) in human cancers such as breast cancer, colon cancer, and acute myelogenous leukemia. However, its clinical significance in cervical cancer has not been established. We explored NR2F6 expression and its clinicopathological significance in early-stage cervical cancer. METHODS: NR2F6 expression in cervical cancer cell lines and cervical cancer tissues was determined by Western blotting, real-time PCR, and immunochemistry (IHC). NR2F6 expression in 189 human early-stage cervical cancer tissue samples was evaluated using IHC. The relevance between NR2F6 expression and early-stage cervical cancer prognosis and clinicopathological features was determined. RESULTS: There was marked NR2F6 mRNA and protein overexpression in the cervical cancer cells and clinical tissues compared with an immortalized squamous cell line and adjacent noncancerous cervical tissues, respectively. In the 189 cervical cancer samples, NR2F6 expression was positively related to International Federation of Gynecology and Obstetrics (FIGO) stage (p = 0.006), squamous cell carcinoma antigen (p = 0.006), vital status (p < 0.001), tumor recurrence (p = 0.001), chemotherapy (p = 0.039), and lymph node metastasis (p < 0.001). Overall and disease-free survival was shorter in patients with early-stage cervical cancer and higher NR2F6 levels than in patients with lower levels of NR2F6. Univariate and multivariate analysis determined that NR2F6 was an independent prognostic factor of survival in early-stage cervical cancer. CONCLUSIONS: Taken together, our findings suggest that high NR2F6 expression predicts pelvic lymph node metastasis, tumor recurrence and poor prognosis in early-stage cervical cancer. NR2F6 might be a novel prognostic biomarker and potential therapeutic target of cervical cancer.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Ganglios Linfáticos/patología , Recurrencia Local de Neoplasia/patología , Receptores de Esteroides/biosíntesis , Neoplasias del Cuello Uterino/patología , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Supervivencia sin Enfermedad , Femenino , Células HeLa , Humanos , Metástasis Linfática/patología , Persona de Mediana Edad , Estadificación de Neoplasias , Receptores de Esteroides/genética , Proteínas Represoras , Neoplasias del Cuello Uterino/mortalidad
18.
Nat Commun ; 7: 12702, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27581363

RESUMEN

Metabolic pathways are reprogrammed in cancer to support cell survival. Here, we report that T-cell acute lymphoblastic leukemia (T-ALL) cells are characterized by increased oxidative phosphorylation and robust ATP production. We demonstrate that ORP4L is expressed in T-ALL but not normal T-cells and its abundance is proportional to cellular ATP. ORP4L acts as an adaptor/scaffold assembling CD3ɛ, Gαq/11 and PLCß3 into a complex that activates PLCß3. PLCß3 catalyzes IP3 production in T-ALL as opposed to PLCγ1 in normal T-cells. Up-regulation of ORP4L thus results in a switch in the enzyme responsible for IP3-induced endoplasmic reticulum Ca(2+) release and oxidative phosphorylation. ORP4L knockdown results in suboptimal bioenergetics, cell death and abrogation of T-ALL engraftment in vivo. In summary, we uncovered a signalling pathway operating specifically in T-ALL cells in which ORP4L mediates G protein-coupled ligand-induced PLCß3 activation, resulting in an increase of mitochondrial respiration for cell survival. Targeting ORP4L might represent a promising approach for T-ALL treatment.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Fosfolipasa C beta/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Receptores de Esteroides/biosíntesis , Adenosina Trifosfato/biosíntesis , Animales , Línea Celular Tumoral , Supervivencia Celular/fisiología , Retículo Endoplásmico/metabolismo , Femenino , Humanos , Células Jurkat , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/metabolismo , Fosforilación Oxidativa , Linfocitos T/metabolismo
19.
BMC Genomics ; 17 Suppl 7: 517, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27557147

RESUMEN

BACKGROUND: In combination with gene expression profiles, the protein interaction network (PIN) constructs a dynamic network that includes multiple functional modules. Previous studies have demonstrated that rifampin can influence drug metabolism by regulating drug-metabolizing enzymes, transporters, and microRNAs (miRNAs). Rifampin induces gene expression, at least in part, by activating the pregnane X receptor (PXR), which induces gene expression; however, the impact of rifampin on global gene regulation has not been examined under the molecular network frameworks. METHODS: In this study, we extracted rifampin-induced significant differentially expressed genes (SDG) based on the gene expression profile. By integrating the SDG and human protein interaction network (HPIN), we constructed the rifampin-regulated protein interaction network (RrPIN). Based on gene expression measurements, we extracted a subnetwork that showed enriched changes in molecular activity. Using the Kyoto Encyclopedia of Genes and Genomes (KEGG), we identified the crucial rifampin-regulated biological pathways and associated genes. In addition, genes targeted by miRNAs that were significantly differentially expressed in the miRNA expression profile were extracted based on the miRNA-gene prediction tools. The miRNA-regulated PIN was further constructed using associated genes and miRNAs. For each miRNA, we further evaluated the potential impact by the gene interaction network using pathway analysis. RESULTS AND DISCCUSSION: We extracted the functional modules, which included 84 genes and 89 interactions, from the RrPIN, and identified 19 key rifampin-response genes that are associated with seven function pathways that include drug response and metabolism, and cancer pathways; many of the pathways were supported by previous studies. In addition, we identified that a set of 6 genes (CAV1, CREBBP, SMAD3, TRAF2, KBKG, and THBS1) functioning as gene hubs in the subnetworks that are regulated by rifampin. It is also suggested that 12 differentially expressed miRNAs were associated with 6 biological pathways. CONCLUSIONS: Our results suggest that rifampin contributes to changes in the expression of genes by regulating key molecules in the protein interaction networks. This study offers valuable insights into rifampin-induced biological mechanisms at the level of miRNAs, genes and proteins.


Asunto(s)
Inactivación Metabólica/genética , Mapas de Interacción de Proteínas/genética , Rifampin/uso terapéutico , Tuberculosis/genética , Antibióticos Antituberculosos/uso terapéutico , Biología Computacional , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/genética , Análisis por Micromatrices , Receptor X de Pregnano , Mapas de Interacción de Proteínas/efectos de los fármacos , Receptores de Esteroides/biosíntesis , Receptores de Esteroides/genética , Tuberculosis/tratamiento farmacológico
20.
Int J Oncol ; 49(2): 639-45, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27279570

RESUMEN

Activation of intestinal human pregnane X receptor (PXR) has recently been proposed as a promising strategy for the chemoprevention of inflammation-induced colon cancer. The present study was aimed at evaluating the effect of rifaximin, a non-absorbable antibiotic, in inhibiting angiogenesis in a model of human colorectal epithelium and investigating the role of PXR in its mechanism of action. Caco-2 cells were treated with rifaximin (0.1, 1.0 and 10.0 µM) in the presence or absence of ketoconazole (10 µM) and assessed for cell proliferation, migration and expression of proliferating cell nuclear antigen (PCNA). The release of vascular endothelial growth factor (VEGF) and nitric oxide (NO), expression of Akt, mechanistic target of rapamycin (mTOR), p38 mitogen activated protein kinases (MAPK), nuclear factor κB (NF-κB) and metalloproteinase-2 and -9 (MMP-2 and -9) were also evaluated. Treatment with rifaximin 0.1, 1.0 and 10.0 µM caused significant and concentration-dependent reduction of cell proliferation, cell migration and PCNA expression in the Caco-2 cells vs. untreated cells. Treatment downregulated VEGF secretion, NO release, VEGFR-2 expression, MMP-2 and MMP-9 expression vs. untreated cells. Rifaximin treatment also resulted in a concentration-dependent decrease in the phosphorylation of Akt, mTOR, p38MAPK and inhibition of hypoxia-inducible factor 1-α (HIF-1α), p70S6K and NF-κB. Ketoconazole (PXR antagonist) treatment inhibited these effects. These findings demonstrated that rifaximin causes PXR-mediated inhibition of angiogenic factors in Caco-2 cell line and may be a promising anticancer tool.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Receptores de Esteroides/biosíntesis , Rifamicinas/administración & dosificación , Antibacterianos/administración & dosificación , Células CACO-2 , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/etiología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/patología , Cetoconazol/administración & dosificación , Proteínas de Neoplasias/biosíntesis , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Receptor X de Pregnano , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Receptores de Esteroides/antagonistas & inhibidores , Rifaximina , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...