Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Hum Reprod ; 16(3): 215-27, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19906824

RESUMEN

Aberrant expression of microRNAs (miRNAs), including miR-21, and alteration of their target genes stability have been associated with cellular transformation and tumorigenesis. We investigated the expression, regulation and function of miR-21 in leiomyomas which develop from myometrial cellular transformation. The results indicated that miR-21 is over-expressed in leiomyomas with specific elevation during the secretory phase of the menstrual cycle and in women who received Depo-Provera and oral contraceptives, but reduced due to GnRHa therapy (P < 0.05). Bioinformatic analysis of microarray gene expression profiles previously obtained from the above cohorts, and myometrial smooth muscle cells (MSMC) and leiomyoma smooth muscle cells (LSMC) treated with GnRHa, transforming growth factor (TGF)-beta and TGF-beta receptor type II (TGF-betaRII) antisense oligomer, indicated that a number of miR-21-predicted target genes were co-expressed and differentially regulated in these cohorts. Gain- and loss-of-function of miR-21 in MSMC, LSMC, transformed LSMC and leiomyosarcoma cell line (SKLM-S1) resulted in differential expression of many genes, including some of the miR-21-predicted/validated target genes, PTEN, PDCD4 and E2F1, and TGF-betaRII, in a cell-specific manner. Gain-of miR-21 function in MSMC and LSMC reduced TGF-beta-induced expression of fibromodulin and TGF-beta-induced factor (P < 0.05), and moderately altered the rate of cell growth and caspase-3/7 activity in these cells. We concluded that miR-21 is aberrantly expressed and hormonally regulated in leiomyomas where, through functional interaction with ovarian steroids and the TGF-beta signaling pathway, either directly or indirectly regulates a number of genes whose products are critical in leiomyoma growth and regression as well as their potential cellular transformation.


Asunto(s)
Leiomioma/metabolismo , Leiomiosarcoma/metabolismo , MicroARNs/metabolismo , Neoplasias Uterinas/metabolismo , Adulto , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Biología Computacional , Factor de Transcripción E2F1/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/farmacología , Humanos , MicroARNs/genética , Persona de Mediana Edad , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfohidrolasa PTEN/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/farmacología , Células Tumorales Cultivadas
2.
Mol Hum Reprod ; 13(11): 797-806, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17766684

RESUMEN

MicroRNAs (miRNAs), through mRNA degradation or repression, act as key regulator of gene expression. Our aim was to identify specific miRNAs that are expressed in endometrium of women with and without endometriosis. We profiled the expression of 287 miRNAs in paired eutopic and ectopic endometrium and isolated endometrial cells using microarray and validated the expression of selected miRNAs using real-time PCR. On the basis of global normalization, 65 of these miRNAs were identified to be expressed above the threshold levels set during the analysis in the endometrium of women without endometriosis with a progressive decline in expression in paired eutopic and ectopic endometrium. Statistical analysis (ANOVA) identified 48 of these miRNAs as differentially expressed among these tissues and 32 miRNAs between isolated endometrial stromal cell (ESC) and glandular epithelial cell (GEC) (P < 0.05). The expression of hsa-miR20a, hsa-miR21, hsa-miR26a, hsa-miR18a, hsa-miR206, hsa-miR181a and hsa-miR142-5p, predicted to target many genes, including TGF-betaR2, ERalpha, ERbeta and PR, respectively, was validated in these tissues and cells using real-time PCR. Treatment of ESC and GEC with 17beta-estradiol and medroxyprogesterone acetate (10(-8) M) differentially regulated the expression of hsa-miR20a, hsa-miR21 and hsa-miR26a, which in part reversed following co-treatment with ICI-182780 and RU-486 (10(-6) M), respectively (P < 0.05). In conclusion, we provided evidence for the expression of a number of differentially expressed miRNAs in eutopic/ectopic endometrium and isolated endometrial cells, opening up the possibility that aberrant/altered expression of some miRNAs whose expression is regulated by the ovarian steroids may influence the expression of specific target genes with central roles in normal endometrial cellular activities and pathogenesis of endometriosis.


Asunto(s)
Endometriosis/genética , Endometrio/efectos de los fármacos , Perfilación de la Expresión Génica , MicroARNs/genética , Esteroides/farmacología , Adulto , Endometrio/citología , Endometrio/metabolismo , Estradiol/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Acetato de Medroxiprogesterona/farmacología , MicroARNs/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovario/efectos de los fármacos , Ovario/metabolismo , Reacción en Cadena de la Polimerasa
3.
J Reprod Immunol ; 73(2): 118-129, 2007 04.
Artículo en Inglés | MEDLINE | ID: mdl-17010446

RESUMEN

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editors following an investigation by the Office of Research Integrity (ORI) at the Department of Health and Human Services. The investigation confirmed that the data presented has been falsified by the last author.


Asunto(s)
Antibacterianos/farmacología , Anticonceptivos Femeninos/farmacología , Doxiciclina/farmacología , Endometrio/enzimología , Células Epiteliales/enzimología , Estradiol/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Metaloproteinasas de la Matriz/biosíntesis , Acetato de Medroxiprogesterona/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Línea Celular , Anticonceptivos Femeninos/efectos adversos , Relación Dosis-Respuesta a Droga , Endometrio/citología , Endometrio/inmunología , Células Epiteliales/citología , Células Epiteliales/inmunología , Femenino , Regulación Enzimológica de la Expresión Génica/inmunología , Hemorragia/inducido químicamente , Humanos , Metaloproteinasas de la Matriz/inmunología , Acetato de Medroxiprogesterona/efectos adversos , Células del Estroma/citología , Células del Estroma/enzimología , Células del Estroma/inmunología , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Inhibidor Tisular de Metaloproteinasa-1/inmunología , Inhibidor Tisular de Metaloproteinasa-2/biosíntesis , Inhibidor Tisular de Metaloproteinasa-2/inmunología
4.
Reprod Biol Endocrinol ; 5: 34, 2007 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-17716379

RESUMEN

BACKGROUND: Clinical observations indicate that leiomyomas occur more frequently in African Americans compared to other ethnic groups with unknown etiology. To identify the molecular basis for the difference we compared leiomyomas form A. Americans with Caucasians using genomic and proteomic strategies. METHODS: Microarray, realtime PCR, 2D-PAGE, mass spectrometry, Western blotting and immunohistochemistry. RESULTS: Using Affymetrix U133A array and analysis based on P ranking (P < 0.01) 1470 genes were identified as differentially expressed in leiomyomas compared to myometrium regardless of ethnicity. Of these, 268 genes were either over-expressed (177 genes) or under-expressed (91 genes) based on P < 0.01 followed by 2-fold cutoff selection in leiomyomas of A. Americans as compared to Caucasians. Among them, the expression E2F1, RUNX3, EGR3, TBPIP, ECM2, ESM1, THBS1, GAS1, ADAM17, CST6, CST7, FBLN5, ICAM2, EDN1 and COL18 was validated using realtime PCR low-density arrays. 2D PAGE coupled with image analysis identified 332 protein spots of which the density/volume of 31 varied by greater than or equal to 1.5 fold in leiomyomas as compared to myometrium. The density/volume of 34 protein-spots varied by greater than or equal to 1.5 fold (26 increased and 8 decreased) in leiomyomas of A. Americans as compared to Caucasians. Tandem mass spectrometric analysis of 15 protein spots identified several proteins whose transcripts were also identified by microarray, including 14-3-3 beta and mimecan, whose expression was confirmed using western blotting and immunohistochemistry. CONCLUSION: These findings imply that the level rather than the ethnic-specific expression of a number of genes and proteins may account for the difference between leiomyomas and possibly myometrium, in A. Americans and Caucasians. Further study using larger sample size is required to confirm these findings.


Asunto(s)
Negro o Afroamericano/genética , Perfilación de la Expresión Génica , Genómica , Leiomioma/genética , Proteómica , Neoplasias Uterinas/genética , Población Blanca/genética , Adulto , Femenino , Perfilación de la Expresión Génica/métodos , Genómica/métodos , Humanos , Leiomioma/patología , Proteómica/métodos , Neoplasias Uterinas/patología
5.
Reprod Biol Endocrinol ; 5: 35, 2007 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-17718906

RESUMEN

BACKGROUND: Leiomyoma have often been compared to keloids because of their fibrotic characteristic and higher rate of occurrence among African Americans as compared to other ethnic groups. To evaluate such a correlation at molecular level this study comparatively analyzed leiomyomas with keloids, surgical scars and peritoneal adhesions to identify genes that are either commonly and/or individually distinguish these fibrotic disorders despite differences in the nature of their development and growth. METHODS: Microarray gene expression profiling and realtime PCR. RESULTS: The analysis identified 3 to 12% of the genes on the arrays as differentially expressed among these tissues based on P ranking at greater than or equal to 0.005 followed by 2-fold cutoff change selection. Of these genes about 400 genes were identified as differentially expressed in leiomyomas as compared to keloids/incisional scars, and 85 genes as compared to peritoneal adhesions (greater than or equal to 0.01). Functional analysis indicated that the majority of these genes serve as regulators of cell growth (cell cycle/apoptosis), tissue turnover, transcription factors and signal transduction. Of these genes the expression of E2F1, RUNX3, EGR3, TBPIP, ECM-2, ESM1, THBS1, GAS1, ADAM17, CST6, FBLN5, and COL18A was confirmed in these tissues using quantitative realtime PCR based on low-density arrays. CONCLUSION: the results indicated that the molecular feature of leiomyomas is comparable but may be under different tissue-specific regulatory control to those of keloids and differ at the levels rather than tissue-specific expression of selected number of genes functionally regulating cell growth and apoptosis, inflammation, angiogenesis and tissue turnover.


Asunto(s)
Cicatriz/genética , Expresión Génica , Queloide/genética , Leiomioma/genética , Enfermedades Peritoneales/genética , Neoplasias Uterinas/genética , Adulto , Negro o Afroamericano/genética , Cicatriz/epidemiología , Cicatriz/metabolismo , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Genómica , Humanos , Queloide/epidemiología , Queloide/metabolismo , Leiomioma/epidemiología , Leiomioma/metabolismo , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Enfermedades Peritoneales/metabolismo , Reacción en Cadena de la Polimerasa , Prevalencia , Proteómica , ARN Mensajero/metabolismo , Adherencias Tisulares/genética , Adherencias Tisulares/metabolismo , Neoplasias Uterinas/epidemiología , Neoplasias Uterinas/metabolismo
6.
Pharmacotherapy ; 26(3): 333-40, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16503719

RESUMEN

STUDY OBJECTIVE: To investigate the immunomodulatory effects of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) by determining whether atorvastatin alters the production of specific endothelium-derived immunoactive proteins and whether its treatment effects depend on its concentration and/or inhibition of 3-hydroxy-3-methylglutaryl coenzyme A reductase. DESIGN: In vitro study using a multiplexing method for protein measurement. SETTING: University laboratory. MEASUREMENTS AND MAIN RESULTS: Human umbilical vein endothelial cells were cultured to approximately 80% confluence and treated with atorvastatin 1-50 microM alone or with mevalonate for 24 hours. Untreated cells served as controls. Culture-conditioned media were removed and multiplex assayed for protein content of epithelial neutrophil-activating peptide-78, interleukin-8, monocyte chemotactic protein-1, interleukin-6, interleukin-10, fibroblast growth factor, and granulocyte colony stimulating factor. Atorvastatin significantly reduced the production of epithelial neutrophil-activating peptide-78, interleukin-6, interleukin-8, and monocyte chemotactic protein-1 (p<0.001 to p<0.05) in a concentration-dependent manner without affecting basal production of interleukin-10, fibroblast growth factor, and granulocyte colony stimulating factor. The treatment effects of atorvastatin were reversed with concurrent mevalonate therapy. CONCLUSION: By inhibiting 3-hydroxy-3-methylglutaryl coenzyme A reductase, atorvastatin lowered concentrations of several inflammatory molecules derived from basal-state endothelial cells in a concentration-dependent manner. The in vivo importance of these immunomodulatory effects needs further investigation.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Factores Inmunológicos/farmacología , Pirroles/farmacología , Atorvastatina , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CXCL5 , Quimiocinas CXC/metabolismo , Células Endoteliales/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Ácido Mevalónico/farmacología
7.
Endocrinology ; 146(3): 1097-118, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15604209

RESUMEN

Altered expression of the TGF-beta system is recognized to play a central role in various fibrotic disorders, including leiomyoma. In this study we performed microarray analysis to characterize the gene expression profile of leiomyoma and matched myometrial smooth muscle cells (LSMC and MSMC, respectively) in response to the time-dependent action of TGF-beta and, after pretreatment with TGF-beta type II receptor (TGF-beta RII) antisense oligomer-blocking/reducing TGF-beta autocrine/paracrine actions. Unsupervised and supervised assessments of the gene expression values with a false discovery rate selected at P < or = 0.001 identified 310 genes as differentially expressed and regulated in LSMC and MSMC in a cell- and time-dependent manner by TGF-beta. Pretreatment with TGF-beta RII antisense resulted in changes in the expression of many of the 310 genes regulated by TGF-beta, with 54 genes displaying a response to TGF-beta treatment. Comparative analysis of the gene expression profile in TGF-beta RII antisense- and GnRH analog-treated cells indicated that these treatments target the expression of 222 genes in a cell-specific manner. Gene ontology assigned these genes functions as cell cycle regulators, transcription factors, signal transducers, tissue turnover, and apoptosis. We validated the expression and TGF-beta time-dependent regulation of IL-11, TGF-beta-induced factor, TGF-beta-inducible early gene response, early growth response 3, CITED2 (cAMP response element binding protein-binding protein/p300-interacting transactivator with ED-rich tail), Nur77, Runx1, Runx2, p27, p57, growth arrest-specific 1, and G protein-coupled receptor kinase 5 in LSMC and MSMC using real-time PCR. Together, the results provide the first comprehensive assessment of the LSMC and MSMC molecular environment targeted by autocrine/paracrine action of TGF-beta, highlighting potential involvement of specific genes whose products may influence the outcome of leiomyoma growth and fibrotic characteristics by regulating inflammatory response, cell growth, apoptosis, and tissue remodeling.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Regulación de la Expresión Génica , Leiomioma/metabolismo , Miocitos del Músculo Liso/metabolismo , Miometrio/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Apoptosis , Células Cultivadas , Análisis por Conglomerados , Estudios de Cohortes , Medios de Cultivo , Medio de Cultivo Libre de Suero , Femenino , Fibrosis , Eliminación de Gen , Humanos , Inflamación , Oligonucleótidos Antisentido/química , ARN/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Tiempo , Factor de Crecimiento Transformador beta1
8.
Endocrinology ; 146(3): 1074-96, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15604208

RESUMEN

Gene microarray was used to characterize the molecular environment of leiomyoma and matched myometrium during growth and in response to GnRH analog (GnRHa) therapy as well as GnRHa direct action on primary cultures of leiomyoma and myometrial smooth muscle cells (LSMC and MSMC). Unsupervised and supervised analysis of gene expression values and statistical analysis in R programming with a false discovery rate of P < or = 0.02 resulted in identification of 153 and 122 differentially expressed genes in leiomyoma and myometrium in untreated and GnRHa-treated cohorts, respectively. The expression of 170 and 164 genes was affected by GnRHa therapy in these tissues compared with their respective untreated group. GnRHa (0.1 microm), in a time-dependent manner (2, 6, and 12 h), targeted the expression of 281 genes (P < or = 0.005) in LSMC and MSMC, 48 of which genes were found in common with GnRHa-treated tissues. Functional annotations assigned these genes as key regulators of processes involving transcription, translational, signal transduction, structural activities, and apoptosis. We validated the expression of IL-11, early growth response 3, TGF-beta-induced factor, TGF-beta-inducible early gene response, CITED2 (cAMP response element binding protein-binding protein/p300-interacting transactivator with ED-rich tail), Nur77, growth arrest-specific 1, p27, p57, and G protein-coupled receptor kinase 5, representing cytokine, common transcription factors, cell cycle regulators, and signal transduction, at tissue levels and in LSMC and MSMC in response to GnRHa time-dependent action using real-time PCR, Western blotting, and immunohistochemistry. In conclusion, using different, complementary approaches, we characterized leiomyoma and myometrium molecular fingerprints and identified several previously unrecognized genes as targets of GnRHa action, implying that local expression and activation of these genes may represent features differentiating leiomyoma and myometrial environments during growth and GnRHa-induced regression.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/análogos & derivados , Leiomioma/metabolismo , Miometrio/metabolismo , Neoplasias Uterinas/metabolismo , Transporte Activo de Núcleo Celular , Western Blotting , Análisis por Conglomerados , Estudios de Cohortes , ADN Complementario/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Inmunohistoquímica , Modelos Biológicos , Miocitos del Músculo Liso/citología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Análisis de Secuencia por Matrices de Oligonucleótidos , Premenopausia , Procesamiento Proteico-Postraduccional , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares , Receptores de Esteroides , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba
9.
Fertil Steril ; 83(3): 659-65, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15749495

RESUMEN

OBJECTIVE: To determine the relationship between uterine bleeding patterns in levonorgestrel users with endometrial histology and expression of interleukins (IL) IL-13 and IL-15. DESIGN: Prospective observational study. SETTING: Academic research center. PATIENT(S): Questionnaires were sent to patients (n = 578) who had levonorgestrel implants concerning bleeding patterns; 35 of these patients were identified to have regular cycle (n = 13), amenorrhea (n = 8), or metrorrhagia (n = 14). INTERVENTION(S): Endometrial biopsies, serum, histology, and immunostaining. MAIN OUTCOME MEASURE(S): Endometrial histological assessment and immunostaining for IL-13 and IL-15 and for blood levonorgestrel, E2, and progesterone levels by ELISA or RIA. RESULT(S): No correlation was found between circulating levonorgestrel, E2, or progesterone levels with the bleeding patterns, although a trend toward a lower E2 level was observed in patients with amenorrhea who had inactive endometrium. There was a direct correlation between bleeding patterns and endometrial histology, as well as IL-13 and IL-15 expression in patients with regular cycles and metrorrhagia, demonstrating secretory and proliferative endometrium, respectively. Some patients in each group were also identified as demonstrating endometritis. CONCLUSION(S): Endometrial histology may assist directing therapy and subsequently increasing compliance in progestin-only contraceptive users with irregular bleeding who fail to respond to standard therapies. Altered endometrial expression of IL-13 and IL-15, key cytokines in inflammatory and immune cell trafficking, may influence events, leading to irregular bleeding.


Asunto(s)
Amenorrea/metabolismo , Anticonceptivos Sintéticos Orales/uso terapéutico , Interleucina-13/metabolismo , Interleucina-15/metabolismo , Levonorgestrel/uso terapéutico , Menorragia/metabolismo , Amenorrea/patología , Biomarcadores , Biopsia , Anticonceptivos Sintéticos Orales/sangre , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Endometrio/patología , Estradiol/sangre , Femenino , Humanos , Levonorgestrel/sangre , Menorragia/patología , Menstruación/fisiología , Cooperación del Paciente , Progesterona/sangre , Encuestas y Cuestionarios
10.
J Clin Endocrinol Metab ; 88(3): 1350-61, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12629129

RESUMEN

The objective of this study was to further elucidate the role of TGFbeta and GnRH analog (GnRHa) in leiomyoma growth and regression. We examined the expression of Smads, TGFbeta receptor intracellular signaling molecules, in leiomyoma and myometrial smooth muscle cells (LSMC and MSMC), and determined whether TGFbeta and GnRHa differentially regulate their expression and induction in these cells. Using semiquantitative RT-PCR, Western blot analysis, and immunohistochemistry, we demonstrated that leiomyoma, myometrium, LSMC, and MSMC express receptor-activated Smad3, common Smad4, and the inhibitory Smad7 mRNA and protein and showed that TGFbeta1, in a time-dependent manner, transiently induced Smad7 expression, with Smad3 and Smad4 remaining largely unchanged. TGFbeta1 increased the rate of Smad and phosphorylated Smad3 (pSmad3) induction in both cell types. Pretreatment with TGFbeta type II receptor antisense oligonucleotide resulted in a trend toward a lower TGFbeta-induced pSmad3. GnRHa, in a dose- and time-dependent manner, increased the expression of Smad7 mRNA and the rapid induction of Smad3, Smad4, and Smad7 as well as pSmad3, which declined to control values at doses above 1 micro M in MSMC, but not in LSMC. GnRHa-induced pSamd3 was partly inhibited by a GnRH antagonist (antide). We concluded that leiomyoma, myometrium, LSMC, and MSMC express Smads, which are differentially expressed, induced, and activated by TGFbeta and are altered as a result of GnRHa treatment. These results suggest that TGFbeta and GnRHa mediate their actions through cross-talk involving Smads and most likely other signaling pathways that result in leiomyoma growth and regression.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/efectos de los fármacos , Leiomioma/metabolismo , Leuprolida/farmacología , Miocitos del Músculo Liso/metabolismo , Miometrio/metabolismo , Transducción de Señal , Transactivadores/genética , Factor de Crecimiento Transformador beta/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Perfilación de la Expresión Génica , Humanos , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Proteína smad3 , Proteína Smad4 , Proteína smad7
11.
J Clin Endocrinol Metab ; 88(10): 4967-76, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14557482

RESUMEN

Human endometrium expresses TGF-beta and TGF-beta receptors where they regulate several endometrial biological activities implicated in embryo implantation, irregular bleeding, endometriosis, and cancer. In the present study, we determined the expression of Smads, intracellular signals that mediate TGF-beta receptors signals from the cell surface to the nucleus, in the endometrium as well as isolated endometrial epithelial (EEC) and stromal (ESC) cells. We also determined whether TGF-beta regulates the expression Smads and activates Smad3 in these cells and endometrial surface epithelial cell line (HES). Using semiquantitative RT-PCR, Western blot analysis, and immunohistochemistry, we found that endometrium, EEC, ESC, and HES express Smad3, -4, and -7 mRNA and protein and contain phosphorylated Smad3 (pSmad3). Smads and pSmad3 were localized in the epithelial and stromal cells with cytoplasmic/nuclear localization. TGF-beta in a dose- and time-dependent manner increased the expression of Smads mRNA and protein, the rate of pSmad3 activation, and Smad3 translocation into the nucleus in ESC and HES. The effect of TGF-beta on pSmad3 induction was, in part, abrogated by the pretreatment of HES and ESC with TGF-beta type II receptor antisense oligonucleotides. We conclude that human endometrium expresses the necessary components of Smad signaling pathway, whose expression and induction in endometrial epithelial and stromal cells are regulated by TGF-beta.


Asunto(s)
Proteínas de Unión al ADN/genética , Endometrio/fisiología , Células Epiteliales/fisiología , Células del Estroma/fisiología , Transactivadores/genética , Factor de Crecimiento Transformador beta/farmacología , Adulto , Línea Celular , Relación Dosis-Respuesta a Droga , Endometrio/citología , Células Epiteliales/citología , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Humanos , Premenopausia , ARN Mensajero/análisis , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteína smad3 , Proteína Smad4 , Proteína smad7 , Células del Estroma/citología
12.
J Clin Endocrinol Metab ; 89(11): 5549-57, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15531510

RESUMEN

GnRH analog (GnRHa) and TGF-beta act directly on leiomyoma/myometrial smooth muscle cells (LSMCs and MSMCs) regulating diverse activities resulting in leiomyoma growth and regression. Because GnRH and TGF-beta receptor signaling is in part mediated through the MAPK pathway, we determined whether the contribution of MAPK/ERK and transcriptional activation of c-fos and c-jun, result in differential regulation of type I collagen, fibronectin, and plasminogen activator inhibitor 1 (PAI-1) gene expression, whose products are known to influence extracellular matrix turnover, which is critical in leiomyoma growth and GnRHa-induced regression. We found that GnRHa and TGF-beta in a dose- and time-dependent manner increased the level of phosphorylated ERK1/2 (pERK1/2) in LSMCs and MSMCs. GnRHa and TGF-beta increased ERK1/2 nuclear accumulation resulting in differential regulation of c-fos and c-jun mRNA expression via downstream signaling from MAPK kinase (MEK)1/2, because pretreatment with U0126, a synthetic inhibitor of MEK1/2, abolished basal and GnRHa- and TGF-beta-induced pERK1/2 and the expression of c-fos and c-jun. LSMCs and MSMCs also express fibronectin, type I collagen, and PAI-1 mRNA, and GnRHa and TGF-beta altered their expression in a cell-specific manner through MEK1/2. We concluded that GnRHa and TGF-beta acting through a MAPK/ERK pathway and transcriptional activation of c-fos/c-jun results in differential regulation of specific genes whose products may in part influence the outcome of leiomyoma growth and regression.


Asunto(s)
Colágeno Tipo I/genética , Fibronectinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/farmacología , Leiomioma/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocitos del Músculo Liso/metabolismo , Miometrio/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Factor de Crecimiento Transformador beta/farmacología , Neoplasias Uterinas/metabolismo , Activación Enzimática , Femenino , Genes fos , Genes jun , Humanos
13.
Front Biosci ; 7: e91-115, 2002 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11897550

RESUMEN

Whether induced by infection, inflammation, ischemia, and/or surgical injury, peritoneal adhesions are the leading cause of pelvic pain, bowel obstruction and infertility. It is clear that while postsurgical peritoneal wounds heal without adhesions in some patients, others develop severe scarring from seemingly equal procedures; in addition, in the same patient, adhesions can develop at one surgical site and not in another. The mechanisms underlying the predisposition to form adhesions as well as their site specificity are completely unknown. However, a large number of intraperitoneal surgical procedures are performed each day in the USA, and thus many patients are at risk of developing postoperative adhesions. Therefore, understanding of adhesion formation at the molecular level is essential and in the absence of such information, attempts to prevent patients from developing adhesions will remain an empirical process. The unprecedented advancement in molecular biology during the past decade has led to the identification of many biologically active molecules with the potential of regulating inflammatory and immune responses, angiogenesis and tissue remodeling, events that are central to normal peritoneal wound healing and adhesion formation. Although, the insight into their importance in the development of tissue fibrosis has substantially increased, their major roles in peritoneal biological functions and adhesion formation remain at best speculative. This article reviews the clinical implications of adhesions and attempts to highlight some of the key molecules i.e. growth factors, cytokines, chemokines, proteases and extracellular matrix, that are recognized to regulate inflammation, fibrinolysis, angiogenesis, and tissue remodeling, events that are central to peritoneal wound repair and adhesion formation. Finally, the article discusses the potential application and site specific delivery of several active compounds that are developed to alter the local inflammatory and immune response i.e., cytokine/chemokine network, targeted gene delivery and development of a new generation of biomaterials to prevent adhesion formation. Such understanding of peritoneal biology not only assist us to better manage patients with adhesion, but also those with endometriosis and malignant diseases that affect the peritoneal cavity.


Asunto(s)
Enfermedades Peritoneales/etiología , Materiales Biocompatibles/uso terapéutico , Citocinas/fisiología , Fibrinólisis , Sustancias de Crecimiento/fisiología , Humanos , Inflamación/complicaciones , Metaloproteinasas de la Matriz/fisiología , Modelos Biológicos , Neovascularización Patológica , Enfermedades Peritoneales/sangre , Enfermedades Peritoneales/terapia , Peritoneo/cirugía , Complicaciones Posoperatorias , Adherencias Tisulares/sangre , Adherencias Tisulares/etiología , Adherencias Tisulares/terapia , Cicatrización de Heridas
14.
Mol Cell Endocrinol ; 209(1-2): 9-16, 2003 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-14604812

RESUMEN

Gene microarray analysis indicated that several components of the transforming growth factor beta receptor (TGF-betaR) signaling pathway are differentially expressed in leiomyoma and myometrium. To validate the microarray results we evaluated the expression of Smads, intracellular proteins that transmit TGF-betaR signals, in leiomyoma and matched myometrium from untreated women, and women who received gonadotropin releasing hormone analogue (GnRHa) therapy. Semi-quantitative RT-PCR, Western blotting and immunohistochemistry indicate that leiomyoma and myometrium expresses receptor-activated Smad3, common Smad4 and inhibitory Smad7, with elevated expression of Smad3, Smad4 and phosphorylated Smad3 (pSmad3) as well as TGF-betaR type I and type II in leiomyoma compared to myometrium (P<0.05). GnRHa therapy resulted in lowering of TGF-betaRs as well as Smad4 and pSmad3, with concurrent increased in Smad7 expression in both leiomyoma and myometrium compared to untreated group (P<0.05). Immunohistochemically Smads and pSmad3 were localized in cytoplasmic/nuclear compartments of leiomyoma and myometrial smooth muscle cells and connective tissue fibroblasts, with alteration in their intensity (HScores) in GnRHa-treated group. In conclusion, the results indicates that leiomyoma and myometrium express all the components of TGF-betaR and Smads, and GnRHa therapy results in alteration of their expression further supporting the importance of TGF-beta system as key regulator of leiomyoma growth.


Asunto(s)
Leiomioma/tratamiento farmacológico , Leiomioma/metabolismo , Leuprolida/uso terapéutico , Miometrio/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transactivadores/metabolismo , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Leiomioma/genética , Miometrio/química , Miometrio/patología , ARN Mensajero/genética , Receptores de Factores de Crecimiento Transformadores beta/análisis , Proteína smad3 , Proteína Smad4 , Proteína smad7 , Transactivadores/análisis , Transactivadores/genética
15.
J Reprod Immunol ; 56(1-2): 93-110, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12106886

RESUMEN

Interleukins (IL)-13 and IL-15 are novel cytokines and key regulators of immune/inflammatory related responses that are critical in the outcome of various normal biological and associated abnormalities of the endometrium. The present study determined the temporal and spatial expression of IL-13 and IL-15 mRNA and protein in endometrium of normal fertile women throughout the menstrual cycle, and examined whether profiles of their expression differ from endometrium of women with unexplained recurrent spontaneous abortion (RSA). Using quantitative RT-PCR, ELISA and immunohistochemistry we found that IL-13 and IL-15 mRNA and protein are expressed in control endometrium throughout the menstrual cycle and in RSA (cycle days 21-23) with peak expression detected immediately after and prior to onset of menses, and two distinct periods corresponding to late proliferative and the early-mid secretory phases, respectively. The ratio of IL-13:IL-15 expression revealed a predominance in IL-13 expression during late proliferative/early secretory phase and IL-15 during the mid secretory phase. Compared to control endometrium, endometrium of women with RSA expresses elevated levels of IL-13 and IL-15, with IL-13:IL-15 ratio favoring IL-13. The immunoreactive IL-13 and IL-15 were localized primarily in endometrial luminal epithelial cells with an increased intensity in glandular epithelial and stromal cells in RSA. In conclusion, the results indicate that endometrium of normal fertile women expresses IL-13 and IL-15, with a distinct profile during the menstrual cycle and elevated expression in women with RSA. Although the biological significance of IL-13 and IL-15 in human endometrium and their elevated expression in RSA await investigation, these cytokines with distinct biological functions may regulate endometrial inflammatory/immune responses, tissue repair and receptivity for embryo implantation.


Asunto(s)
Aborto Habitual/metabolismo , Endometrio/metabolismo , Interleucina-13/genética , Interleucina-15/genética , Ciclo Menstrual/metabolismo , ARN Mensajero/análisis , Femenino , Humanos , Inmunohistoquímica , Interleucina-13/análisis , Interleucina-13/biosíntesis , Interleucina-15/análisis , Interleucina-15/biosíntesis , Embarazo
16.
Reprod Biol Endocrinol ; 1: 125, 2003 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-14678567

RESUMEN

Gonadotropin releasing hormone analogues (GnRHa) are often used to regress endometriosis implants and prevent premature luteinizing hormone surges in women undergoing controlled ovarian stimulation. In addition to GnRH central action, the expression of GnRH and receptors in the endometrium implies an autocrine/paracrine role for GnRH and an additional site of action for GnRHa. To further examine the direct action of GnRH (Leuprolide acetate) in the endometrium, we determined the effect of GnRH on endometrial stromal (ESC) and endometrial surface epithelial (HES) cells expression and activation of Smads (Smad3, -4 and -7), intracellular signals activated by transforming growth factor beta (TGF-beta), a key cytokine expressed in the endometrium. The results show that GnRH (0.1 microM) increased the expression of inhibitory Smad7 mRNA in HES with a limited effect on ESC, while moderately increasing the common Smad4 and Smad7 protein levels in these cells (P < 0.05). GnRH in a dose--(0.01 to 10 microM) and time--(5 to 30 min) dependent manner decreased the rate of Smad3 activation (phospho-Smad3, pSmad3), and altered Smad3 cellular distribution in both cell types. Pretreatment with Antide (GnRH antagonist) resulted in further suppression of Smad3 induced by GnRH, with Antide inhibition of pSmad3 in ESC. Furthermore, co-treatment of the cells with GnRH + TGF-beta, or pretreatment with TGF-beta type II receptor antisense to block TGF-beta autocrine/paracrine action, in part inhibited TGF-beta activated Smad3. In conclusion, the results indicate that GnRH acts directly on the endometrial cells altering the expression and activation of Smads, a mechanism that could lead to interruption of TGF-beta receptor signaling mediated through this pathway in the endometrium.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Endometrio/efectos de los fármacos , Hormona Liberadora de Gonadotropina/agonistas , Leuprolida/farmacología , Transactivadores/biosíntesis , Adulto , Células Cultivadas/efectos de los fármacos , Medio de Cultivo Libre de Suero/farmacología , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta a Droga , Endometrio/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Antagonistas de Hormonas/farmacología , Humanos , Oligodesoxirribonucleótidos Antisentido/farmacología , Oligopéptidos/farmacología , Proteínas Serina-Treonina Quinasas , ARN Mensajero/biosíntesis , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/genética , Proteína smad3 , Proteína Smad4 , Proteína smad7 , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Transactivadores/genética , Factor de Crecimiento Transformador beta/farmacología
17.
Fertil Steril ; 80(3): 564-70, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12969699

RESUMEN

OBJECTIVE: To determine the expression of matrix metalloproteinase (MMP-26) and tissue inhibitor of MMP (TIMP) in the endometrium of women with normal menstrual cycles compared with users of levonorgestrel implants. DESIGN: Prospective observational study. SETTING: Academic research center. PATIENT(S): Fifty patients with normal menstrual cycles who requested permanent surgical sterilization (tubal ligation) and 35 users of levonorgestrel implants. INTERVENTION(S): Endometrial biopsy. MAIN OUTCOME MEASURE(S): Expression of MMP-26, TIMP-3, and TIMP-4 by immunohistochemistry and semiquantitative analysis of staining intensity by using the H score. RESULT(S): Endometrium from women with a normal menstrual cycle and users of levonorgestrel implants expresses MMP-26, TIMP-3, and TIMP-4. These substances are present in various types of endometrial cells; expression is strongest in surface and glandular epithelial cells, followed by vascular endothelial and endometrial stromal cells. Inflammatory and immune-related cells also stained strongly for MMP-26 and TIMPs. Semiquantitative analysis of the staining intensity of endometrial epithelial and stromal cells indicated that expression of MMP-26, TIMP-3, and TIMP-4 peaks during the early to mid-luteal phase. Expression of MMP-26 is elevated in users of levonorgestrel implants who experienced irregular uterine bleeding. CONCLUSION(S): Endometrial expression of MMP-26 and TIMP-4 is present throughout the menstrual cycle and is elevated during the early to mid-luteal phase in normally cycling women. Further elevations in MMP-26 are seen in users of levonorgestrel implants who experience irregular uterine bleeding. These substances thus seem to play a role in hormonal regulation and endometrial tissue remodeling.


Asunto(s)
Anticonceptivos Femeninos/efectos adversos , Endometrio/metabolismo , Levonorgestrel/efectos adversos , Metaloproteinasas de la Matriz/metabolismo , Ciclo Menstrual/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Hemorragia Uterina/metabolismo , Adolescente , Adulto , Anticonceptivos Femeninos/administración & dosificación , Implantes de Medicamentos , Femenino , Humanos , Inmunohistoquímica/métodos , Levonorgestrel/administración & dosificación , Metaloproteinasas de la Matriz Secretadas , Estudios Prospectivos , Valores de Referencia , Coloración y Etiquetado , Hemorragia Uterina/inducido químicamente , Inhibidor Tisular de Metaloproteinasa-4
18.
Fertil Steril ; 79(5): 1149-53, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12738510

RESUMEN

OBJECTIVE: To assess the expression and cellular distribution of pregnancy-associated plasma protein-A (PAPP-A) and major basic protein (MBP) in human ovarian tissue during the menstrual cycle. DESIGN: Ovarian tissues (n = 50) and corpora lutea (n = 18) were obtained from patients undergoing hysterectomy/oophorectomy for benign conditions and tissue sections were immunostained for MBP and PAPP-A. SETTING: University medical center. INTERVENTION(S): Immunostaining of tissue sections using antibodies to PAPP-A and MBP. MAIN OUTCOME MEASURE(S): Microscopic evaluation to assess the presence, distribution, and cellular co-localization of MBP and PAPP-A and to describe any variations in their expression during the menstrual cycle. RESULT(S): Major basic protein (MBP) is found in several ovarian cell types throughout the menstrual cycle. The MBP immunostaining of ovarian follicles varied depending on the size, with primordial follicles staining in the ooplasm with a lack of staining in the granulosa and theca cells. In the intermediate/mature follicles, MBP was immunolocalized in theca, but not in granulosa cells except in the mature follicles. Pregnancy-associated plasma protein-A (PAPP-A) was immunolocalized in primordial follicle ooplasm, theca externa of intermediate/mature follicles, and in granulosa cells with increased intensity as luteinization progressed. The luteal tissue is the major site of MBP and PAPP-A with highest intensity found during the midluteal phase associated with both small and large luteal cells. CONCLUSION(S): The expression and distinct pattern of MBP and PAPP-A cellular localization in human ovarian tissue during folliculogenesis and in luteal tissue suggest that their individual and combined actions in a cell specific fashion may play a role in growth and differentiation of theca, granulosa, and luteal cells.


Asunto(s)
Proteínas Sanguíneas/análisis , Cuerpo Lúteo/química , Ciclo Menstrual/metabolismo , Ovario/química , Proteína Plasmática A Asociada al Embarazo/análisis , Ribonucleasas , Proteínas en los Gránulos del Eosinófilo , Femenino , Humanos , Inmunohistoquímica , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/análisis
19.
Eur Cytokine Netw ; 13(1): 128-33, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11956032

RESUMEN

AIMS: To determine whether granulocyte macrophage-colony stimulating factor (GM-CSF) production by neuronal precursor (NT2) cells can be regulated by IL-1beta and TNF-alpha. BACKGROUND: We have previously demonstrated GM-CSF expression by neurons of the developing human brain, as well as by NT2 cells. IL-1beta and TNF-alpha upregulate GM-CSF production in glial cells, but GM-CSF regulation in neurons is as yet undefined. We hypothesized that IL-1beta and TNF-alpha would increase GM-CSF mRNA and protein production in NT2 cells. METHODS: The effect of IL-1beta and TNF-alpha on GM-CSF production was assessed by dose response (0 to 2,000 U/ml), and time course (0 to 48 hours incubation) experiments. GM-CSF mRNA and protein production were assessed by quantitative RT-PCR and by ELISA. The effect of these cytokines on cell turnover was determined by BrdU incorporation. RESULTS: IL-1beta increased GM-CSF mRNA and protein expression by NT2 cells. This effect was time and dose dependent, and the effective dose ranging from (20-200 U/ml). TNF-alpha increased GM-CSF mRNA expression to a lesser extent than did IL-1beta (maximal stimulation at 200 U/ml), and a minimal increase in net protein accumulation was noted. Neither cytokine increased NT2 cell turnover. CONCLUSIONS: IL-1beta and TNF-alpha both increase GM-CSF mRNA expression by NT2 cells, but only IL-1beta increases net GM-CSF protein accumulation.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Interleucina-1/farmacología , Neuronas/efectos de los fármacos , Células Madre/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Línea Celular , Células Cultivadas , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Neuronas/metabolismo , ARN Mensajero/biosíntesis , Células Madre/citología , Células Madre/metabolismo
20.
J Soc Gynecol Investig ; 10(1): 21-6, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12517589

RESUMEN

Because gonadotropin releasing hormone analogue (GnRHa) therapy often causes leiomyoma regression, in part through alteration of growth factor and receptor expression, we determined whether GnRHa therapy alters the expression of extracellular signal-regulated kinase (ERK) and focal adhesion kinase (FAK), which are linked to intracellular signaling pathways activated by ovarian steroids, growth factors, and adhesion molecules. Leiomyoma and matched unaffected myometrium were collected from women who received GnRHa therapy (n = 5) and untreated women (n = 10). We determined the expression of ERK1, ERK2, FAK, phosphorylated ERK (pERK1/2), and pFAK using Western blotting and immunohistochemical analysis. Leiomyoma and myometrium expressed ERK1 (44 kD), ERK2 (42 kD), and FAK (125 kD) at variable levels with increased ERK2, pERK, and FAK expression in leiomyoma. We found that GnRHa therapy resulted in a noticeable decrease in ERK2 and FAK, with significant reduction in pERK1/2 and low or undetectable levels of pFAK in both leiomyoma and myometrium compared with the untreated group (P <.05). Immunohistochemically ERK1, ERK2, FAK, pERK1/2, and pFAK were localized in smooth muscle cells and connective tissue fibroblasts in GnRHa-treated and untreated leiomyoma and myometrium, with considerable reduction in their intensity as indicated by HScore in GnRHa-treated tissues. The data provide further evidence that leiomyoma regression induced by GnRHa is mediated in part through a mechanism involving suppression of signal transduction pathways involving growth factors or ovarian steroid and adhesion molecules.


Asunto(s)
Leiomioma/enzimología , Leuprolida/uso terapéutico , Proteínas Quinasas Activadas por Mitógenos/análisis , Proteínas Tirosina Quinasas/análisis , Transducción de Señal/efectos de los fármacos , Neoplasias Uterinas/enzimología , Antineoplásicos Hormonales/uso terapéutico , Western Blotting , Femenino , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Humanos , Inmunohistoquímica , Leiomioma/tratamiento farmacológico , Proteína Quinasa 1 Activada por Mitógenos/análisis , Proteína Quinasa 3 Activada por Mitógenos , Premenopausia , Neoplasias Uterinas/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA