Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
Oncogene ; 40(13): 2407-2421, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33664454

RESUMEN

Metastatic castration-resistant prostate cancer (mCRPC) is a malignant and lethal disease caused by relapse after androgen-deprivation (ADT) therapy. Since enzalutamide is innovated and approved by US FDA as a new treatment option for mCRPC patients, drug resistance for enzalutamide is a critical issue during clinical usage. Although several underlying mechanisms causing enzalutamide resistance were previously identified, most of them revealed that drug resistant cells are still highly addicted to androgen and AR functions. Due to the numerous physical functions of AR in men, innovated AR-independent therapy might alleviate enzalutamide resistance and prevent production of adverse side effects. Here, we have identified that yes-associated protein 1 (YAP1) is overexpressed in enzalutamide-resistant (EnzaR) cells. Furthermore, enzalutamide-induced YAP1 expression is mediated through the function of chicken ovalbumin upstream promoter transcription factor 2 (COUP-TFII) at the transcriptional and the post-transcriptional levels. Functional analyses reveal that YAP1 positively regulates numerous genes related to cancer stemness and lipid metabolism and interacts with COUP-TFII to form a transcriptional complex. More importantly, YAP1 inhibitor attenuates the growth and cancer stemness of EnzaR cells in vitro and in vivo. Finally, YAP1, COUP-TFII, and miR-21 are detected in the extracellular vesicles (EVs) isolated from EnzaR cells and sera of patients. In addition, treatment with EnzaR-EVs induces the abilities of cancer stemness, lipid metabolism and enzalutamide resistance in its parental cells. Taken together, these results suggest that YAP1 might be a crucial factor involved in the development of enzalutamide resistance and can be an alternative therapeutic target in prostate cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Benzamidas/administración & dosificación , Factor de Transcripción COUP II/genética , MicroARNs/genética , Nitrilos/administración & dosificación , Feniltiohidantoína/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Factores de Transcripción/genética , Anciano , Animales , Benzamidas/efectos adversos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Xenoinjertos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Persona de Mediana Edad , Células Madre Neoplásicas/efectos de los fármacos , Nitrilos/efectos adversos , Feniltiohidantoína/efectos adversos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Señalizadoras YAP
3.
Reproduction ; 161(1): 11-19, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33112285

RESUMEN

Endometriosis is a common gynecological disease in reproductive-age women. Although the hormone-dependent therapy is the first line treatment for endometriosis, it is not a curative regimen and associated with severe side-effects, which significantly decrease the life quality of affected individuals. To seek a target for treatment of endometriosis, we focused on plasma membrane proteins that are elevated in ectopic cells and exert beneficial effects in cell growth and survival. We performed bioinformatics analysis and identified the neurotrophic receptor tyrosine kinase 2 (NTRK2) as a potential candidate for treatment. The expression levels of NTRK2 were markedly upregulated in the lesions of clinical specimen as well as in the mouse endometriotic-like lesion. Mechanistic investigation demonstrated that upregulation of NTRK2 is induced by hypoxia in a hypoxia-inducible factor 1 alpha-dependent manner. Knockdown of NTRK2 or administration of ANA-12, a selective antagonist of NTRK2, significantly induced endometriotic stromal cells death, suggesting it may be a potential therapeutic agent. In vivo study using surgery-induced endometriosis mice model showed ANA-12 (1.5 mg/kg body weight) treatment induced apoptosis of endometriotic cells and caused the regression of ectopic lesions. Taken together, our findings suggest a possible mechanism responsible for the aberrant expression of NTRK2 in endometriotic lesions and this may be involved in the pathogenesis of endometriosis.


Asunto(s)
Endometriosis/tratamiento farmacológico , Glicoproteínas de Membrana/metabolismo , ARN Interferente Pequeño/uso terapéutico , Receptor trkB/metabolismo , Animales , Coristoma/metabolismo , Evaluación Preclínica de Medicamentos , Endometriosis/metabolismo , Femenino , Humanos , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Cultivo Primario de Células , ARN Interferente Pequeño/farmacología , Receptor trkB/antagonistas & inhibidores , Células del Estroma/metabolismo
4.
Theranostics ; 9(3): 620-632, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30809297

RESUMEN

Rationale: Endometriosis is a highly prevalent gynecological disease in women of reproductive age that markedly reduces life quality and fertility. Unfortunately, there is no cure for this disease, which highlights that more efforts are needed to investigate the underlying mechanism for designing novel therapeutic regimens. This study aims to investigate druggable membrane receptors distinctively expressed in endometriotic cells. Methods: Bioinformatic analysis of public databases was employed to identify potential druggable candidates. Normal endometrial tissues and ectopic endometriotic lesions were obtained for the determination of target genes. Primary endometrial and endometriotic stromal cells as well as two different mouse models of endometriosis were used to characterize molecular mechanisms and therapeutic outcomes of endometriosis, respectively. Results: Anthrax toxin receptor 2 (ANTXR2) mRNA and protein are upregulated in the endometriotic specimens. Elevation of ANTXR2 promotes endometriotic cell adhesion, proliferation, and angiogenesis. Furthermore, hypoxia is the driving force for ANTXR2 upregulation via altering histone modification of ANTXR2 promoter by reducing the repressive mark, histone H3 lysine 27 (H3K27) trimethylation, and increasing the active mark, H3K4 trimethylation. Activation of ANTXR2 signaling leads to increased Yes-associated protein 1 (YAP1) nuclear translocation and transcriptional activity, which contributes to numerous pathological processes of endometriosis. Pharmacological blocking of ANTXR2 signaling not only prevents endometriotic lesion development but also causes the regression of established lesion. Conclusion: Taken together, we have identified a novel target that contributes to the disease pathogenesis of endometriosis and provided a potential therapeutic regimen to treat it.


Asunto(s)
Endometriosis/patología , Endometriosis/terapia , Receptores de Péptidos/antagonistas & inhibidores , Receptores de Péptidos/análisis , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Hipoxia , Ratones Endogámicos C57BL , Receptores de Péptidos/genética
5.
Reprod Sci ; 26(6): 734-744, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30092712

RESUMEN

Endometriosis is a common gynecological disease, which is defined as the growth of endometrial tissues outside the uterine cavity. It often causes dysmenorrhea, dyspareunia, chronic pelvic pain, and infertility in reproductive-age women. However, the pathogenesis of endometriosis remains largely unclear. Since our previous study revealed that ectopic endometriotic stromal cells experience greater hypoxic stress than their eutopic counterparts, we aim to investigate whether the metabolic properties are changed in the ectopic endometriotic stromal cell when compared to its eutopic counterpart. Here, we found the expression of pyruvate dehydrogenase kinase 1 (PDK1), a critical enzyme in regulating glucose metabolism, was increased in ectopic stromal cells. Molecular characterization reveals that overexpression of PDK1 is induced by hypoxia through transcriptional regulation. Upregulation of PDK1 in ectopic endometriotic stromal cells was accompanied by increases in lactate production and oxygen consumption rate when compared to eutopic endometrial stromal cells. Furthermore, our data showed that inhibition of PDK1 activity by treatment with dichloroacetate inhibits the lactate production and oxygen consumption rate of ectopic stromal cells. In addition, hypoxia-induced PDK1 expression prevented cells from H2O2- and low nutrient-induced cell death. These data indicate that ectopic endometriotic cells may adapt to hypoxic microenvironment via upregulating PDK1 and reprogramming metabolism, which provides a survival advantage in the hostile peritoneal microenvironment.


Asunto(s)
Apoptosis/fisiología , Hipoxia de la Célula/fisiología , Endometriosis/patología , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/fisiología , Células del Estroma/fisiología , Apoptosis/efectos de los fármacos , Células Cultivadas , Femenino , Expresión Génica/fisiología , Regulación de la Expresión Génica , Glucólisis/fisiología , Humanos , Peróxido de Hidrógeno/farmacología , Ácido Láctico/biosíntesis , Consumo de Oxígeno/fisiología
6.
J Endocr Soc ; 2(7): 765-778, 2018 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-30151432

RESUMEN

CONTEXT: Progesterone (P) resistance is a hallmark of endometriosis, but the underlying mechanism(s) for loss of P sensitivity leading to lesion establishment remains poorly understood. OBJECTIVE: To evaluate the association between Notch-1 signaling activation and P resistance in the progression of endometriosis. DESIGN: Case control study; archived formalin-fixed, paraffin-embedded tissues. SETTING: University hospitals (United States, Taiwan). PATIENTS: Women with endometriosis; human endometrial stromal cell line (HESC). INTERVENTION: Eutopic endometria (EU) and ectopic lesions (ECs) were collected from surgically diagnosed patients. Archived tissue sections of EU and ECs were identified. HESCs were treated with N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT) and valproic acid (VPA) to, respectively, suppress and induce Notch-1 activation. OUTCOME MEASURES: Tissues were analyzed for Notch Intra-Cellular Domain 1 (NICD1) and progesterone receptor (PGR) protein expression by immunohistochemistry and for transcript levels of NICD1 target genes HES1, PGR, and PGR-B by quantitative reverse transcription polymerase chain reaction. DAPT- or VPA-treated HESCs with and without P cotreatment were evaluated for cell numbers and for PGR, HES1, and PGR target gene DKK1 transcript levels. RESULTS: Nuclear-localized stromal NICD1 protein levels were inversely associated with those of total PGR in EU and ECs. Stromal ECs displayed higher HES1 and lower total PGR and PGR-B transcript levels than EU. In HESCs, DAPT reduction of NICD1 decreased cell numbers and increased PGR transcript and nuclear PGR protein levels and, with P cotreatment, maintained P sensitivity. Conversely, VPA induction of NICD1 decreased PGR transcript levels and, with P cotreatment, abrogated P-induced DKK1 and maintained HES1 transcript levels. CONCLUSIONS: Aberrant Notch-1 activation is associated with decreased PGR that contributes to P resistance in endometriosis.

7.
Hum Reprod ; 33(8): 1517-1527, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29982401

RESUMEN

STUDY QUESTION: How does hypoxia-mediated downregulation of chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) promote angiogenesis in endometriosis? SUMMARY ANSWER: Suppression of COUP-TFII by hypoxia stimulates angiogenesis through induction of angiogenin (ANG). WHAT IS KNOWN ALREADY: The level of COUP-TFII is downregulated in endometriotic tissues, and downregulation of COUP-TFII contributes to the development of endometriosis. STUDY DESIGN, SIZE, DURATION: Twenty-seven patients of reproductive age with endometriosis were recruited in this study. Eutopic endometrial and ectopic endometriotic stromal cells were isolated, cultured and subjected to various treatments. PARTICIPANTS/MATERIALS, SETTING, METHODS: Microarray hybridization, quantitative RT-PCR, and Western blot were used to detect gene expression in normal and endometriotic samples. A luciferase reporter assay and chromatin immunoprecipitation in normoxia- or hypoxia-treated primary cultures of human endometrial stromal cells were performed. Tube formation analysis was performed using primary human umbilical vein endothelial cells (HUVECs). MAIN RESULTS AND THE ROLE OF CHANCE: Protein level of COUP-TFII was downregulated by hypoxia (P < 0.05, normoxia versus hypoxia). Loss of COUP-TFII increased the angiogenic capacity of endometrial stromal cells (P < 0.05, COUP-TFII knockdown versus knockdown control). A novel COUP-TFII target gene, ANG, was identified through microarray analysis. Chromatin immunoprecipitation and promoter activity assays demonstrated that the ANG promoter was bound and suppressed by COUP-TFII (P < 0.05, COUP-TFII overexpression versus empty vector). The levels of ANG mRNA and protein were elevated in ectopic endometriotic stromal cells and negatively correlated with COUP-TFII (P < 0.05, endometrial versus endometriotic tissues/stromal cells). Both knockdown and forced-expression of COUP-TFII further demonstrated that ANG expression and ANG-mediated angiogenic activity were negatively regulated by COUP-TFII (P < 0.05, COUP-TFII knockdown versus knockdown control, and COUP-TFII overexpression versus empty vector). LIMITATIONS, REASONS FOR CAUTION: This study was conducted in primary human endometrial stromal cell cultures and HUVECs, therefore, may not fully reflect the situation in vivo. LARGE SCALE DATA: The raw data were submitted to Gene Expression Omnibus (GSE107469). WIDER IMPLICATIONS OF THE FINDINGS: This is the first study to highlight that the aberrant expression of ANG in endometriotic lesions is mediated by hypoxia-suppressed COUP-TFII expression, which reveals an as yet unidentified molecular pathogenesis of endometriosis. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by research grants (MOST 105-2314-B-006-059-MY3 to M.H.W. and MOST 104-2320-B-006-036-MY3 to S.J.T.) from the Ministry of Science and Technology, Taiwan. The authors declare that there is no conflict of interest.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Endometriosis/metabolismo , Endometrio/irrigación sanguínea , Endometrio/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Patológica , Comunicación Paracrina , Ribonucleasa Pancreática/metabolismo , Células del Estroma/metabolismo , Factor de Transcripción COUP II/genética , Estudios de Casos y Controles , Hipoxia de la Célula , Células Cultivadas , Microambiente Celular , Endometriosis/genética , Endometriosis/patología , Endometrio/patología , Femenino , Humanos , Cultivo Primario de Células , Ribonucleasa Pancreática/genética , Transducción de Señal , Células del Estroma/patología
8.
J Pathol ; 242(4): 476-487, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28608501

RESUMEN

Endometriosis is a highly prevalent gynaecological disease that severely reduces women's health and quality of life. Ectopic endometriotic lesions have evolved mechanisms to survive in the hypoxic peritoneal microenvironment by regulating the expression of a significant subset of genes. However, the master regulator controlling these genes remains to be characterized. Herein, by using bioinformatics analysis and experimental verification, we identified yes-associated protein 1 (YAP1) as a master regulator of endometriosis. Nuclear localization and transcriptional activity of YAP1 were up-regulated by hypoxia via down-regulation of LATS1, a kinase that inactivates YAP1. Disruption of hypoxia-induced YAP1 signalling by siRNA knockdown or inhibitor treatment abolished critical biological processes involved in endometriosis development such as steroidogenesis, angiogenesis, inflammation, migration, innervation, and cell proliferation. Treatment with a YAP1 inhibitor caused the regression of endometriotic lesions without affecting maternal fertility or the growth rate of offspring in the mouse model of endometriosis. Taken together, we identify hypoxia/LATS1/YAP1 as a novel pathway for the pathogenesis of endometriosis and demonstrate that targeting YAP1 might be an alternative approach to treat endometriosis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Endometriosis/tratamiento farmacológico , Fertilidad/efectos de los fármacos , Fosfoproteínas/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Proteínas de Ciclo Celular , Hipoxia de la Célula/genética , Hipoxia de la Célula/fisiología , Biología Computacional/métodos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Endometriosis/etiología , Endometriosis/genética , Endometriosis/metabolismo , Femenino , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Estudio de Asociación del Genoma Completo , Humanos , Ratones Endogámicos C57BL , Terapia Molecular Dirigida/métodos , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , Fosfoproteínas/fisiología , Porfirinas/farmacología , Porfirinas/uso terapéutico , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Células del Estroma/metabolismo , Factores de Transcripción , Transcripción Genética , Verteporfina , Proteínas Señalizadoras YAP
9.
J Endocrinol ; 234(1): R53-R65, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28455430

RESUMEN

Endocrine is an important and tightly regulated system for maintaining body homeostasis. Endocrine glands produce hormones, which are released into blood stream to guide the target cells responding to all sorts of stimulations. For maintaining body homeostasis, the secretion and activity of a particular hormone needs to be adjusted in responding to environmental challenges such as changes in nutritional status or chronic stress. Hypoxia, a status caused by reduced oxygen availability or imbalance of oxygen consumption/supply in an organ or within a cell, is a stress that affects many physiological and pathological processes. Hypoxic stress in endocrine organs is especially critical because endocrine glands control body homeostasis. Local hypoxia affects not only the particular gland but also the downstream cells/organs regulated by hormones secreted from this gland. Hypoxia-inducible factors (HIFs) are transcription factors that function as master regulators of oxygen homeostasis. Recent studies report that aberrant expression of HIFs in endocrine organs may result in the development and/or progression of diseases including diabetes, endometriosis, infertility and cancers. In this article, we will review recent findings in HIF-mediated endocrine organ dysfunction and the systemic syndromes caused by these disorders.


Asunto(s)
Hipoxia/metabolismo , Oxígeno/metabolismo , Factores de Transcripción/metabolismo , Animales , Regulación de la Expresión Génica/fisiología , Homeostasis/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA