Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Reprod Sci ; 30(2): 544-559, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35732928

RESUMEN

The alterations in myometrial biology during labor are not well understood. The myometrium is the contractile portion of the uterus and contributes to labor, a process that may be regulated by the steroid hormone progesterone. Thus, human myometrial tissues from term pregnant in-active-labor (TIL) and term pregnant not-in-labor (TNIL) subjects were used for genome-wide analyses to elucidate potential future preventive or therapeutic targets involved in the regulation of labor. Using myometrial tissues directly subjected to RNA sequencing (RNA-seq), progesterone receptor (PGR) chromatin immunoprecipitation sequencing (ChIP-seq), and histone modification ChIP-seq, we profiled genome-wide changes associated with gene expression in myometrial smooth muscle tissue in vivo. In TIL myometrium, PGR predominantly occupied promoter regions, including the classical progesterone response element, whereas it bound mainly to intergenic regions in TNIL myometrial tissue. Differential binding analysis uncovered over 1700 differential PGR-bound sites between TIL and TNIL, with 1361 sites gained and 428 lost in labor. Functional analysis identified multiple pathways involved in cAMP-mediated signaling enriched in labor. A three-way integration of the data for ChIP-seq, RNA-seq, and active histone marks uncovered the following genes associated with PGR binding, transcriptional activation, and altered mRNA levels: ATP11A, CBX7, and TNS1. In vitro studies showed that ATP11A, CBX7, and TNS1 are progesterone responsive. We speculate that these genes may contribute to the contractile phenotype of the myometrium during various stages of labor. In conclusion, we provide novel labor-associated genome-wide events and PGR-target genes that can serve as targets for future mechanistic studies.


Asunto(s)
Trabajo de Parto , Progesterona , Embarazo , Femenino , Humanos , Progesterona/metabolismo , Miometrio/metabolismo , Estudio de Asociación del Genoma Completo , Trabajo de Parto/genética , Trabajo de Parto/metabolismo , Unión Proteica , Complejo Represivo Polycomb 1/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(47): e2208886119, 2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36375056

RESUMEN

Uterine leiomyoma is the most common tumor in women and causes severe morbidity in 15 to 30% of reproductive-age women. Epidemiological studies consistently indicate a correlation between leiomyoma development and exposure to endocrine-disrupting chemical phthalates, especially di-(2-ethylhexyl) phthalate (DEHP); however, the underlying mechanisms are unknown. Here, among the most commonly encountered phthalate metabolites, we found the strongest association between the urine levels of mono(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP), the principal DEHP metabolite, and the risk of uterine leiomyoma diagnosis (n = 712 patients). The treatment of primary leiomyoma and smooth muscle cells (n = 29) with various mixtures of phthalate metabolites, at concentrations equivalent to those detected in urine samples, significantly increased cell viability and decreased apoptosis. MEHHP had the strongest effects on both cell viability and apoptosis. MEHHP increased cellular tryptophan and kynurenine levels strikingly and induced the expression of the tryptophan transporters SLC7A5 and SLC7A8, as well as, tryptophan 2,3-dioxygenase (TDO2), the key enzyme catalyzing the conversion of tryptophan to kynurenine that is the endogenous ligand of aryl hydrocarbon receptor (AHR). MEHHP stimulated nuclear localization of AHR and up-regulated the expression of CYP1A1 and CYP1B1, two prototype targets of AHR. siRNA knockdown or pharmacological inhibition of SLC7A5/SLC7A8, TDO2, or AHR abolished MEHHP-mediated effects on leiomyoma cell survival. These findings indicate that MEHHP promotes leiomyoma cell survival by activating the tryptophan-kynurenine-AHR pathway. This study pinpoints MEHHP exposure as a high-risk factor for leiomyoma growth, uncovers a mechanism by which exposure to environmental phthalate impacts leiomyoma pathogenesis, and may lead to the development of novel druggable targets.


Asunto(s)
Dietilhexil Ftalato , Contaminantes Ambientales , Leiomioma , Ácidos Ftálicos , Humanos , Femenino , Dietilhexil Ftalato/toxicidad , Dietilhexil Ftalato/orina , Quinurenina , Triptófano , Supervivencia Celular , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Transportador de Aminoácidos Neutros Grandes 1 , Exposición a Riesgos Ambientales/efectos adversos , Leiomioma/inducido químicamente , Leiomioma/orina
3.
Reprod Sci ; 29(3): 743-749, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35064560

RESUMEN

Uterine leiomyomas (fibroids) are common benign tumors in women. The tryptophan metabolism through the kynurenine pathway plays important roles in tumorigenesis in general. Leiomyomas expressing mutated mediator complex subunit 12 (mut-MED12) were reported to contain significantly decreased tryptophan levels; the underlying mechanism and the role of the tryptophan metabolism-kynurenine pathway in leiomyoma tumorigenesis, however, remain unknown. We here assessed the expression and regulation of the key enzymes that metabolize tryptophan. Among these, the tissue mRNA levels of tryptophan 2,3-dioxygenase (TDO2), the rate limiting enzyme of tryptophan metabolism through the kynurenine pathway, was 36-fold higher in mut-MED12 compared to adjacent myometrium (P < 0.0001), and 14-fold higher compared to wild type (wt)-MED12 leiomyoma (P < 0.05). The mRNA levels of other tryptophan metabolizing enzymes, IDO1 and IDO2, were low and not significantly different, suggesting that TDO2 is the key enzyme responsible for reduced tryptophan levels in mut-MED12 leiomyoma. R5020 and medroxyprogesterone acetate (MPA), two progesterone agonists, regulated TDO2 gene expression in primary myometrial and leiomyoma cells expressing wt-MED12; however, this effect was absent or blunted in leiomyoma cells expressing G44D mut-MED12. These data suggest that MED12 mutation may alter progesterone-mediated TDO2 expression in leiomyoma, leading to lower levels of tryptophan in mut-MED12 leiomyoma. This highlights that fibroids can vary widely in their response to progesterone as a result of mutation status and provides some insight for understanding the effect of tryptophan-kynurenine pathway on leiomyoma tumorigenesis and identifying targeted interventions for fibroids based on their distinct molecular signatures.


Asunto(s)
Leiomioma/enzimología , Complejo Mediador/genética , Triptófano Oxigenasa/metabolismo , Adulto , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Mutación , Progestinas/farmacología , Células Tumorales Cultivadas
4.
Stem Cell Reports ; 16(9): 2099-2106, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34388365

RESUMEN

Uterine leiomyoma (LM) is the most common tumor in women. Via its receptor (PGR) expressed in differentiated LM cells, progesterone stimulates paracrine signaling that induces proliferation of PGR-deficient LM stem cells (LSCs). Antiprogestins shrink LM but tumors regrow after treatment cessation possibly due to persisting LSCs. Using sorted primary LM cell populations, we found that the PGR gene locus and its target cistrome are hypermethylated in LSCs, inhibiting the expression of genes critical for progesterone-induced LSC differentiation. PGR knockdown shifted the transcriptome of total LM cells toward LSCs and increased global DNA methylation by regulating TET methylcytosine dioxygenases. DNA methylation inhibitor 5'-Aza activated PGR signaling, stimulated LSC differentiation, and synergized with antiprogestin to reduce tumor size in vivo. Taken together, targeting the feedback loop between DNA methylation and progesterone signaling may accelerate the depletion of LSCs through rapid differentiation and sensitize LM to antiprogestin therapy, thus preventing tumor regrowth.


Asunto(s)
Biomarcadores de Tumor , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Leiomioma/etiología , Células Madre Neoplásicas/metabolismo , Receptores de Progesterona/genética , Sitios de Unión , Secuencia de Consenso , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunofenotipificación , Leiomioma/tratamiento farmacológico , Leiomioma/metabolismo , Leiomioma/patología , Modelos Biológicos , Motivos de Nucleótidos , Unión Proteica , Receptores de Progesterona/metabolismo
5.
Female Pelvic Med Reconstr Surg ; 27(8): 521-526, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34261104

RESUMEN

OBJECTIVE: The aims of this study were to evaluate the biomolecular properties of vaginal and perineal granulation tissue in postpartum women and assess the potential impact of vaginal estrogen application. METHODS: We prospectively identified women referred to a subspecialty peripartum clinic between September 2016 and April 2018 who developed symptomatic perineal or vaginal granulation tissue. As part of routine clinical care, granulation tissue was excised from each participant by a urogynecologist and subjected to RNA extraction, real-time quantitative polymerase chain reaction, histologic evaluation, and immunohistochemistry. Serum steroid hormone levels were measured. Comparisons were made between participants who used topical vaginal estradiol (E2) and those who did not (non-E2 controls). RESULTS: Sixteen postpartum women were recruited for this pilot study. More than 30% of patients (n = 5, 31%) had used topical vaginal estradiol (E2) during their postpartum recovery. Histological appearance of granulation tissue evaluated by hematoxylin and eosin staining was similar in women treated with vaginal E2 and non-E2 controls. Both estrogen receptor α (ERα) and ERß mRNA and ERα protein were readily detectable in the granulation tissue of E2-treated women. Although not statistically significant, participants who used topical E2 developed granulation tissue that exhibited local estrogen-responsive gene upregulation. Serum levels of estrone, E2, dehydroepiandrosterone, progesterone, and testosterone did not differ between vaginal E2-treated patients and controls. CONCLUSIONS: Estrogen receptor α seems to be the predominant receptor mediating estrogen action in postpartum perineal and vaginal granulation tissue. Vaginal E2 use does not seem to affect serum levels of estrone, E2, dehydroepiandrosterone, progesterone, and testosterone in postpartum women.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Tejido de Granulación/efectos de los fármacos , Vagina/patología , Administración Tópica , Estradiol/administración & dosificación , Receptor alfa de Estrógeno , Estrógenos/administración & dosificación , Femenino , Humanos , Proyectos Piloto , Periodo Posparto
6.
Adipocyte ; 10(1): 310-321, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34085602

RESUMEN

Objectives Adipocytes and adipocyte lipid metabolism are closely related with obesity and type 2 diabetes, but the molecular mechanism still needs further investigation. The aim of this study is to discover the adipocyte genes and pathways involved in obesity and type 2 diabetes using bioinformatics analysis.Methods The GSE27951 gene expression profile was obtained. Software and online tools (STRING, Cytoscape, BioGPS, CTD, and FunRich) were used to identify core genes.21 human subcutaneous adipose samples, with 10 from type 2 diabetic patients and 11 from normal controls, were included in these analyses.Results 184 differentially expressed genes (DEGs) including 42 up-regulated genes and 142 down-regulated genes were found to be enriched in metabolism, receptor activity, collagen type IV and glutamine biosynthesis I pathway by using the enrichment analysis. Seven hub genes were identified from the PPI network using various software (Cytoscape, STRING, BioGPS, and CTD). Four core genes (COL4A2, ACACB, GLUL, and CD36) were found to be highly expressed in subcutaneous adipose tissue of obese patients accompanying type 2 diabetes.Conclusion COL4A2, ACACB, GLUL and CD36 might be the core molecular biomarkers of obesity in patients with or without type 2 diabetes.


Asunto(s)
Acetil-CoA Carboxilasa/genética , Antígenos CD36/genética , Colágeno Tipo IV/genética , Diabetes Mellitus Tipo 2/genética , Glutamato-Amoníaco Ligasa/genética , Obesidad/genética , Biomarcadores/análisis , Biología Computacional , Perfilación de la Expresión Génica , Humanos , Programas Informáticos
7.
Fertil Steril ; 114(6): 1339-1349, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32892998

RESUMEN

OBJECTIVE: To investigate the functional interaction between the Wnt/ß-catenin and protein kinase B (Akt) pathways in leiomyoma stem cells (LSC). DESIGN: Laboratory study. SETTING: Research laboratory. PATIENT(S): Premenopausal women (n = 36; age range: 28 to 49 years) undergoing hysterectomy or myomectomy for leiomyoma. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Gene expression, protein phosphorylation, and cell proliferation. RESULT(S): Cells from human leiomyoma tissues were sorted by fluorescence-activated cell sorting (FACS) into three populations: LSC, intermediate cells (LIC), and differentiated cells (LDC) with the function of the Wnt/ß-catenin and Akt signaling pathways in leiomyoma cells evaluated using real-time quantitative polymerase chain reaction and immunoblot analyses. The Wnt/ß-catenin signaling pathway components were differentially expressed in each leiomyoma cell population. WNT4 was distinctly overexpressed in LIC, and its receptor FZD6 was primarily expressed in LSC. WNT4 stimulated Akt phosphorylation, activated ß-catenin, and increased primary leiomyoma cell proliferation. These stimulatory effects were abolished by cotreatment with the Akt inhibitor, MK-2206. WNT4 up-regulated the expression of pro-proliferative genes, c-Myc and cyclin D1, specifically in LSC; this was also abrogated by Akt inhibition. CONCLUSION(S): Our data suggest that WNT4 regulates LSC proliferation via Akt-dependent ß-catenin activation, representing a key step toward a better understanding of LSC regulation and potentially novel therapeutic targets.


Asunto(s)
Leiomioma/enzimología , Células Madre Neoplásicas/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Uterinas/enzimología , Proteína Wnt4/metabolismo , Adulto , Proliferación Celular , Activación Enzimática , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Leiomioma/genética , Leiomioma/mortalidad , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Fosforilación , Esferoides Celulares , Células Tumorales Cultivadas , Neoplasias Uterinas/genética , Neoplasias Uterinas/patología , Vía de Señalización Wnt , Proteína Wnt4/genética
8.
Endocrinology ; 161(10)2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32812024

RESUMEN

Uterine leiomyoma (LM) is the most common tumor in women and can cause severe morbidity. Leiomyoma growth requires the maintenance and proliferation of a stem cell population. Dysregulated deoxyribonucleic acid (DNA) methylation has been reported in LM, but its role in LM stem cell regulation remains unclear. Here, we fluorescence-activated cell sorting (FACS)-sorted cells from human LM tissues into 3 populations: LM stem cell-like cells (LSC, 5%), LM intermediate cells (LIC, 7%), and differentiated LM cells (LDC, 88%), and we analyzed the transcriptome and epigenetic landscape of LM cells at different differentiation stages. Leiomyoma stem cell-like cells harbored a unique methylome, with 8862 differentially methylated regions compared to LIC and 9444 compared to LDC, most of which were hypermethylated. Consistent with global hypermethylation, transcript levels of TET1 and TET3 methylcytosine dioxygenases were lower in LSC. Integrative analyses revealed an inverse relationship between methylation and gene expression changes during LSC differentiation. In LSC, hypermethylation suppressed the genes important for myometrium- and LM-associated functions, including muscle contraction and hormone action, to maintain stemness. The hypomethylating drug, 5'-Aza, stimulated LSC differentiation, depleting the stem cell population and inhibiting tumor initiation. Our data suggest that DNA methylation maintains the pool of LSC, which is critical for the regeneration of LM tumors.


Asunto(s)
Azacitidina/farmacología , Diferenciación Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Leiomioma/patología , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Uterinas/patología , Adulto , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Azacitidina/administración & dosificación , Recuento de Células , Células Cultivadas , Femenino , Humanos , Leiomioma/tratamiento farmacológico , Ratones , Ratones SCID , Ratones Transgénicos , Persona de Mediana Edad , Mifepristona/administración & dosificación , Mifepristona/farmacología , Terapia Molecular Dirigida , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Neoplasias Uterinas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
9.
PLoS One ; 15(1): e0226885, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31910207

RESUMEN

Although more than 1 in 4 men develop symptomatic inguinal hernia during their lifetime, the molecular mechanism behind inguinal hernia remains unknown. Here, we explored the protein-protein interaction network built on known inguinal hernia-causative genes to identify essential and common downstream proteins for inguinal hernia formation. We discovered that PIK3R1, PTPN11, TGFBR1, CDC42, SOS1, and KRAS were the most essential inguinal hernia-causative proteins and UBC, GRB2, CTNNB1, HSP90AA1, CBL, PLCG1, and CRK were listed as the most commonly-involved downstream proteins. In addition, the transmembrane receptor protein tyrosine kinase signaling pathway was the most frequently found inguinal hernia-related pathway. Our in silico approach was able to uncover a novel molecular mechanism underlying inguinal hernia formation by identifying inguinal hernia-related essential proteins and potential common downstream proteins of inguinal hernia-causative proteins.


Asunto(s)
Redes Reguladoras de Genes , Hernia Inguinal/metabolismo , Mapeo de Interacción de Proteínas/métodos , Análisis por Conglomerados , Simulación por Computador , Predisposición Genética a la Enfermedad , Humanos , Masculino
10.
J Clin Endocrinol Metab ; 104(12): 6256-6264, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31504629

RESUMEN

CONTEXT: Uterine leiomyoma (fibroids) are the most common tumors in women. Recently, perilipin-2 (PLIN2) was identified as a critical target gene of the progesterone receptor; however, its function in the pathogenesis of fibroids is unknown. OBJECTIVE: To determine the function of PLIN2 in leiomyoma cells. DESIGN: Tissue and primary cells from leiomyoma and myometrium were analyzed. PLIN2 function in leiomyoma was assessed using small interfering RNA. RNA-sequencing was performed to identify genome-wide effects of PLIN2 depletion. Metabolic activity was measured using the Seahorse XF96 analyzer. Real-time quantitative PCR and immunoblotting were also performed. SETTING: Laboratory. PATIENTS OR OTHER PARTICIPANTS: Forty-one premenopausal women undergoing surgery for fibroids. MAIN OUTCOME MEASURES: Gene expression, oxygen consumption rate (OCR), extracellular acidification rate (ECAR), and cell proliferation. RESULTS: PLIN2 gene expression was 2.4-fold lower in leiomyoma compared with adjacent myometrium, suggesting a link between PLIN2 deficiency and fibroids. A total of 3877 genes were differentially expressed after PLIN2 knockdown. Gene ontology analysis identified metabolism as the second-highest biological process affected by PLIN2 depletion. OCR (mitochondrial respiration) and ECAR (glycolysis) were significantly upregulated after PLIN2 knockdown; PLIN2-depleted cells had a greater basal metabolic activity and higher metabolic stress response. Cell proliferation was also significantly increased after PLIN2 knockdown. CONCLUSIONS: PLIN2 depletion increases mitochondrial respiration and glycolysis, suggesting that PLIN2 is a critical regulator of metabolic function in leiomyoma cells. PLIN2 deficiency also reprograms leiomyoma cells to a proproliferative phenotype. These findings introduce metabolomics as an area to explore to better understand leiomyoma tumorigenesis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Leiomioma/patología , Miometrio/patología , Perilipina-2/metabolismo , Progesterona/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias Uterinas/patología , Adulto , Biomarcadores de Tumor/genética , Estudios de Casos y Controles , Proliferación Celular , Femenino , Estudios de Seguimiento , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Leiomioma/genética , Leiomioma/metabolismo , Metaboloma , Miometrio/metabolismo , Perilipina-2/antagonistas & inhibidores , Perilipina-2/genética , Pronóstico , ARN Interferente Pequeño , Receptores de Progesterona/genética , Transducción de Señal , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo
11.
Biomed Res Int ; 2019: 6341967, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30881993

RESUMEN

BACKGROUND: Colon cancer is a heterogeneous disease, differing in clinical symptoms, epigenetics, and prognosis for each individual patient. Identifying the core genes is important for early diagnoses and it provides a more precise method for treating colon cancer. MATERIALS AND METHODS: In this study, we wanted to pinpoint these core genes so we obtained GSE101502 microRNA profiles from the GEO database, which resulted in 17 differential expressed microRNAs that were identified by GEO2R analysis. Then, 875 upregulated and 2920 downregulated target genes were predicted by FunRich. GO and KEGG pathway were used to do enrich analysis. RESULTS: GO analysis indicated that upregulated genes were significantly enriched in the regulation of cell communication and signaling and in nervous system development, while the downregulated genes were significantly enriched in nervous system development and regulation of transcription from the RNA polymerase II promoter. KEGG pathway analysis suggested that the upregulated genes were enriched in axon guidance, MAPK signaling pathway, and endocytosis, while the downregulated genes existed in pathways in cancer, focal adhesion, and PI3K-Akt signaling pathway. The top four molecules including 82 hub genes were identified from the PPI network and involved in endocytosis, spliceosome, TGF-beta signaling pathway, and lysosome. Finally, NUDT21, GNB1, CLINT1, and COL1A2 core gene were selected due to their correlation with the prognosis of IIA stage colon cancer. CONCLUSION: this study suggested that NUDT21, GNB1, CLINT1, and COL1A2 might be the core genes for colon cancer that play an important role in the development and prognosis of IIA stage colon cancer.


Asunto(s)
Neoplasias del Colon/genética , Biología Computacional , MicroARNs/genética , Proteínas de Neoplasias/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Colágeno Tipo I/genética , Neoplasias del Colon/patología , Bases de Datos Genéticas , Supervivencia sin Enfermedad , Subunidades beta de la Proteína de Unión al GTP/genética , Regulación Neoplásica de la Expresión Génica/genética , Ontología de Genes , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico
12.
Oncogene ; 38(15): 2722-2735, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30538295

RESUMEN

Progesterone and its receptor, PR, are essential for uterine leiomyoma (LM, a.k.a., fibroid) tumorigenesis, but the underlying cellular and molecular mechanisms remain unclear. The receptor activator of NF-κB (RANKL) was recently identified as a novel progesterone/PR-responsive gene that plays an important role in promoting LM growth. Here, we used RANKL as a representative gene to investigate how steroid hormone, genetic, and epigenetic signals are integrated to regulate LM stem cell (LSC) function. We demonstrated that RANKL specifically upregulates LSC proliferation through activation of Cyclin D1. RANKL gene transcription was robustly induced by the progesterone agonist R5020, leading to a dramatically higher RANKL expression in LM compared to adjacent myometrial (MM) tissue. MethylCap-Seq revealed a differentially methylated region (DMR) adjacent to the distal PR-binding site (PRBS) 87 kb upstream of the RANKL transcription start site. Hypermethylation of the DMR inhibited recruitment of PR to the adjacent PRBS. Luciferase assays indicated that the DMR and distal PRBS constitute a novel RANKL distal regulatory element that actively regulates RANKL expression. Furthermore, MED12 physically interacts with PR in LM tissue. The interaction between MED12 and PR, binding of PR and MED12 to PRBS, and RANKL gene expression are significantly higher in LM containing a distinct MED12 mutation (G44D) than in LM with wild-type MED12. In summary, our findings suggest that DNA methylation and MED12 mutation together constitute a complex regulatory network that affects progesterone/PR-mediated RANKL gene expression, with an important role in activating stem cell proliferation and fibroid tumor development.


Asunto(s)
Proliferación Celular/genética , Metilación de ADN/genética , Leiomioma/genética , Complejo Mediador/genética , Ligando RANK/genética , Receptores de Progesterona/genética , Células Madre/patología , Neoplasias Uterinas/genética , Adulto , Proliferación Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Leiomioma/tratamiento farmacológico , Persona de Mediana Edad , Progesterona/genética , Promegestona/farmacología , Sitio de Iniciación de la Transcripción/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Neoplasias Uterinas/tratamiento farmacológico
13.
Lab Invest ; 98(12): 1575-1587, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30206313

RESUMEN

Cellular senecence is an important biologic endpoint. Naturally occuring (aging) senescence is common in uterine leiomyoma (ULM). AKT is one of major pathways in promoting ULM growth and survival. Inactivation of AKT by MK2206 in ULM resulted in stress-induced senescence in vitro. Study of the senescent phenotypes and molecular changes in ULM may greatly facilitate the understanding of the tumor biology and potential clinical therapy for this common disease associated with high morbidity. To study senescence in a model system that closely resembles primary ULM in vivo, we applied an ex vivo model of three-dimensional (3D) spheroid culture system which maintained the molecular and cellular characteristics of primary ULM and matched myometrium as seen in vivo. Gene expression profiling done on ULM induced to undergo replication (passaging) or stress-induced (MK2206) senescence revealed that ROS and hypoxic-related genes were upregulated in the two types of senescences. Overexpression of two selected genes, WIPI1 and SLITKR4, induced cellular senescence in ULM spheroids. Additionally, administration of ABT263 (a BH3 mimetic) effectively reduced the senescent cells induced in ULM spheroids. This study identified novel genes associated with senescence in ULM and demonstrated a BH3 mimetic to act as a senolytic to remove senescent cells.


Asunto(s)
Compuestos de Anilina/uso terapéutico , Antineoplásicos/uso terapéutico , Senescencia Celular , Leiomioma/metabolismo , Sulfonamidas/uso terapéutico , Neoplasias Uterinas/metabolismo , Adulto , Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Técnicas de Cultivo , Femenino , Compuestos Heterocíclicos con 3 Anillos , Humanos , Leiomioma/tratamiento farmacológico , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Esferoides Celulares , Estrés Fisiológico , Sulfonamidas/farmacología , Transcriptoma , Neoplasias Uterinas/tratamiento farmacológico
14.
J Clin Endocrinol Metab ; 103(5): 1842-1849, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29741640

RESUMEN

Context: Uterine leiomyomas are the most common type of gynecologic tumor in women. Objective: To determine the role of the cytokine receptor activator of nuclear factor κ-Β ligand (RANKL); its receptor, receptor activator of nuclear factor κ-Β (RANK); and the RANKL/RANK pathway inhibitor RANK-Fc in leiomyoma growth. Design: Messenger RNA (mRNA) or protein levels of RANKL, RANK, and proliferation markers cyclin D1 and Ki67 were assessed in various leiomyoma tissues and cell populations. Human xenograft experiments were performed to determine the effects of RANK-Fc on leiomyoma growth in vivo. Setting: Research laboratory. Patients: Twenty-four regularly cycling premenopausal women (age 28 to 49 years) who were not receiving hormone therapy. Interventions: None. Main Outcome Measure: Tumor growth in a murine xenograft model following targeting of the RANKL/RANK pathway with RANK-Fc. Results: RANKL mRNA levels in leiomyoma were significantly higher than those in myometrial tissues. The highest RANK levels were found in the leiomyoma stem cell population, which is deficient in progesterone receptor (PR). Conversely, the highest RANKL levels were found in the PR-rich leiomyoma intermediate cell (LIC) population. R5020, a PR agonist, specifically increased RANKL expression in LICs. RANK-Fc blocked RANKL-induced expression of the proliferative gene cyclin D1. Treatment with RANK-Fc also significantly decreased tumor growth in vivo and diminished the expression of proliferation marker Ki67 in tumors (P < 0.01; n = 4). Conclusions: Treatment with the RANKL/RANK pathway inhibitor RANK-Fc significantly decreased human leiomyoma cell proliferation and tumor growth. This suggests that the RANKL/RANK pathway could serve as a potential target for the prevention and treatment of uterine leiomyoma.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Leiomioma/patología , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/genética , Proteínas Recombinantes de Fusión/farmacología , Neoplasias Uterinas/patología , Adulto , Estudios de Casos y Controles , Células Cultivadas , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Leiomioma/genética , Persona de Mediana Edad , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Neoplasias Uterinas/genética
15.
Reprod Health ; 15(1): 69, 2018 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-29688866

RESUMEN

OBJECTIVE: The aim of this study was to systematically review the available clinical trials examining male infertility after inguinal hernias were repaired using mesh procedures. METHODS: The Cochrane Library, PubMed, Embase, Web of Science, and Chinese Biomedical Medicine Database were investigated. The Jada score was used to evaluate the quality of the studies, "Oxford Centre for Evidence-based Medicine-Levels of Evidence" was used to assess the level of the trials, and descriptive analysis was used to evaluate the studies. RESULTS: Twenty nine related trials with a total of 36,552 patients were investigated, including seven randomized controlled trials (RCTs) with 616 patients and 10 clinical trials (1230 patients) with mesh or non-mesh repairs. The Jada score showed that there were six high quality RCTs and one low quality RCT. Levels of evidence determined from the Oxford Centre for Evidence-based Medicine further demonstrated that those six high quality RCTs also had high levels of evidence. It was found that serum testosterone, LH, and FSH levels declined in the laparoscopic group compared to the open group; however, the testicular volume only slightly increased without statistical significance. Testicular and sexual functions remained unchanged after both laparoscopic transabdominal preperitoneal hernia repair (TAPP) and totally extra-peritoneal repair (TEP). We also compared the different meshes used post-surgeries. VyproII/Timesh lightweight mesh had a diminished effect on sperm motility compared to Marlex heavyweight mesh after a one-year follow-up, but there was no effect after 3 years. Additionally, various open hernia repair procedures (Lichtenstein, mesh plug method, posterior pre-peritoneal mesh repair, and anterior tension-free repair) did not cause infertility. CONCLUSIONS: This systematic review suggests that hernia repair with mesh either in an open or a laparoscopic procedure has no significant effect on male fertility.


Asunto(s)
Hernia Inguinal/cirugía , Herniorrafia/efectos adversos , Infertilidad Masculina/etiología , Mallas Quirúrgicas/efectos adversos , Ensayos Clínicos como Asunto , Humanos , Laparoscopía , Masculino , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...