Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biol Open ; 6(6): 723-731, 2017 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-28483980

RESUMO

The adherens junction associated protein 1 (AJAP1, aka shrew-1) is presumably a type-I transmembrane protein localizing and interacting with the E-cadherin-catenin complex. In various tumors, AJAP1 expression is reduced or lost, including hepatocellular and esophageal squamous cell carcinoma, and glial-derived tumors. The aberrant expression of AJAP1 is associated with alterations in cell migration, invasion, increased tumor growth, and tumor vascularization, suggesting AJAP1 as a putative tumor suppressor. We show that AJAP1 attenuates sprouting angiogenesis by reducing endothelial migration and invasion capacities. Further, we show for the first time that endogenous AJAP1 is associated with the microtubule cytoskeleton. This linkage is independent from cell confluency and stable during angiogenic sprouting in vitro Our work suggests that AJAP1 is a putative negative regulator of angiogenesis, reducing cell migration and invasion by interfering with the microtubule network. Based on our results and those of other authors, we suggest AJAP1 as a novel tumor suppressor and diagnostic marker.

2.
Biol Open ; 5(11): 1607-1619, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27870635

RESUMO

Shrew-1, also called AJAP1, is a transmembrane protein associated with E-cadherin-mediated adherence junctions and a putative tumor suppressor. Apart from its interaction with ß-catenin and involvement in E-cadherin internalization, little structure or function information exists. Here we explored shrew-1 expression during postnatal differentiation of mammary gland as a model system. Immunohistological analyses with antibodies against either the extracellular or the cytoplasmic domains of shrew-1 consistently revealed the expression of full-length shrew-1 in myoepithelial cells, but only part of it in luminal cells. While shrew-1 localization remained unaltered in myoepithelial cells, nuclear localization occurred in luminal cells during lactation. Based on these observations, we identified two unknown shrew-1 transcript variants encoding N-terminally truncated proteins. The smallest shrew-1 protein lacks the extracellular domain and is most likely the only variant present in luminal cells. RNA analyses of human tissues confirmed that the novel transcript variants of shrew-1 exist in vivo and exhibit a differential tissue expression profile. We conclude that our findings are essential for the understanding and interpretation of future functional and interactome analyses of shrew-1 variants.

3.
Fertil Steril ; 106(2): 378-85, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27041028

RESUMO

OBJECTIVE: To study the expression and function of syndecan-4 in endometriosis. DESIGN: Histopathological investigation of eutopic endometrium and experimental laboratory study on a cell line derived from epithelial endometriotic cells (12Z). SETTING: University hospital laboratory. PATIENT(S): One hundred six women (62 controls/44 endometriosis) from the IVF center of Münster University Hospital aged 23-44 undergoing Pipelle biopsy and diagnostic exploratory laparoscopy. INTERVENTION(S): Eutopic endometrial tissue was investigated by immunohistochemistry for the expression of syndecan-4. The human endometriotic cell line 12Z was transiently transfected with syndecan-4 small interfering RNA and investigated for changes in cell behavior. MAIN OUTCOME MEASURE(S): Syndecan-4 expression in eutopic endometrium was evaluated immunohistochemically in endometrial glands and stroma. Scoring results were correlated with the stages of the menstrual cycle and presence or absence of endometriosis. Quantitative polymerase chain reaction was used to measure syndecan-4-dependent expression changes of MMP2, MMP3, MMP9, Rac1, and ATF2. Altered cell behavior was monitored by matrigel invasion assays and cell viability assays. RESULT(S): Syndecan-4 expression was significantly higher in the glands and stroma of patients with endometriosis compared with controls, whereas no menstrual cycle-dependent expression was observed. In 12Z cells, syndecan-4 depletion did not affect cell viability but resulted in a significantly reduced matrigel invasiveness and reduced expression of the small GTPase Rac1, the transcription factor ATF-2, and MMP3. CONCLUSION(S): The upregulation of syndecan-4 in the eutopic endometrium of endometriosis patients may facilitate the pathogenetic process by promoting invasive cell growth via Rac1, MMP3, and ATF-2.


Assuntos
Movimento Celular , Endometriose/metabolismo , Sindecana-4/metabolismo , Fator 2 Ativador da Transcrição/metabolismo , Adulto , Estudos de Casos e Controles , Linhagem Celular , Endometriose/genética , Endometriose/patologia , Feminino , Humanos , Metaloproteinase 3 da Matriz/metabolismo , Fenótipo , Interferência de RNA , Transdução de Sinais , Sindecana-4/genética , Transfecção , Regulação para Cima , Adulto Jovem , Proteínas rac1 de Ligação ao GTP/metabolismo
4.
Reprod Biomed Online ; 32(4): 434-45, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26854065

RESUMO

Endometriosis is characterized by growth of endometrial tissue at ectopic locations. Down-regulation of microRNA miR-200b is observed in endometriosis and malignant disease, driving tumour cells towards an invasive state by enhancing epithelial-to-mesenchymal transition (EMT). miR-200b up-regulation may inhibit EMT and invasive growth in endometriosis. To study its functional impact on the immortalized endometriotic cell line 12Z, the stromal cell line ST-T1b, and primary endometriotic stroma cells, a transient transfection approach with microRNA precursors was employed. Expression of bioinformatically predicted targets of miR-200b was analysed by qPCR. The cellular phenotype was monitored by Matrigel invasion assays, digital-holographic video microscopy and flow cytometry. qPCR revealed significant down-regulation of ZEB1 (P < 0.05) and ZEB2 (P < 0.01) and an increase in E-cadherin (P < 0.01). miR-200b overexpression decreased invasiveness (P < 0.0001) and cell motility (P < 0.05). In contrast, cell proliferation (P < 0.0001) and the stemness-associated side population phenotype (P < 0.01) were enhanced following miR-200b transfection. These properties were possibly due to up-regulation of the pluripotency-associated transcription factor KLF4 (P < 0.05) and require attention when considering therapeutic strategies. In conclusion, up-regulation of miR-200b reverts EMT, emerging as a potential therapeutic approach to inhibit endometriotic cell motility and invasiveness.


Assuntos
Endometriose/genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição Kruppel-Like/genética , MicroRNAs/fisiologia , Proteínas Repressoras/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Linhagem Celular , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo , Endometriose/patologia , Feminino , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/fisiologia , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/fisiologia , Proteínas Repressoras/metabolismo , Proteínas Repressoras/fisiologia , Células Estromais/metabolismo , Regulação para Cima , Homeobox 2 de Ligação a E-box com Dedos de Zinco , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/fisiologia
5.
Proc Natl Acad Sci U S A ; 112(31): 9716-21, 2015 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-26199416

RESUMO

Endometriosis is a debilitating, estrogen-dependent, progesterone-resistant, inflammatory gynecological disease of reproductive age women. Two major clinical symptoms of endometriosis are chronic intolerable pelvic pain and subfertility or infertility, which profoundly affect the quality of life in women. Current hormonal therapies to induce a hypoestrogenic state are unsuccessful because of undesirable side effects, reproductive health concerns, and failure to prevent recurrence of disease. There is a fundamental need to identify nonestrogen or nonsteroidal targets for the treatment of endometriosis. Peritoneal fluid concentrations of prostaglandin E2 (PGE2) are higher in women with endometriosis, and this increased PGE2 plays important role in survival and growth of endometriosis lesions. The objective of the present study was to determine the effects of pharmacological inhibition of PGE2 receptors, EP2 and EP4, on molecular and cellular aspects of the pathogenesis of endometriosis and associated clinical symptoms. Using human fluorescent endometriotic cell lines and chimeric mouse model as preclinical testing platform, our results, to our knowledge for the first time, indicate that selective inhibition of EP2/EP4: (i) decreases growth and survival of endometriosis lesions; (ii) decreases angiogenesis and innervation of endometriosis lesions; (iii) suppresses proinflammatory state of dorsal root ganglia neurons to decrease pelvic pain; (iv) decreases proinflammatory, estrogen-dominant, and progesterone-resistant molecular environment of the endometrium and endometriosis lesions; and (v) restores endometrial functional receptivity through multiple mechanisms. Our novel findings provide a molecular and preclinical basis to formulate long-term nonestrogen or nonsteroidal therapy for endometriosis.


Assuntos
Endometriose/tratamento farmacológico , Endometriose/patologia , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Compostos de Bifenilo/farmacologia , Compostos de Bifenilo/uso terapêutico , Caspase 3/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Endométrio/irrigação sanguínea , Endométrio/patologia , Estrogênios/biossíntese , Feminino , Humanos , Inflamação/patologia , Camundongos , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Dor Pélvica/tratamento farmacológico , Dor Pélvica/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Progesterona/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esteroides/uso terapêutico , Xantonas/farmacologia , Xantonas/uso terapêutico
6.
Mol Cell Endocrinol ; 409: 51-8, 2015 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-25843056

RESUMO

Endometriosis is an inflammatory gynecological disease of reproductive-age women. The prevalence of endometriosis is 5-10% in reproductive-age women. Modern medical treatments are directed to inhibit the action of estrogen in endometriotic cells. However, hormonal therapies targeting estrogen can be prescribed only for a short time because of their undesirable side effects. Recent studies from our laboratory, using human endometriotic epithelial cell line 12Z and stromal cell line 22B derived from red lesion, discovered that selective inhibition of prostaglandin E2 (PGE2) receptors EP2 and EP4 inhibits adhesion, invasion, growth, and survival of 12Z and 22B cells by modulating integrins, MMPs and TIMPs, cell cycle, survival, and intrinsic apoptotic pathways, suggesting multiple epigenetic mechanisms. The novel findings of the present study indicate that selective pharmacological inhibition of EP2 and EP4: (i) decreases expression of DNMT3a, DNMT3b, H3K9me3, H3K27me3, SUV39H1, HP1a, H3K27, EZH2, JMJD2a, HDAC1, HDAC3, MeCP2, CoREST and Sin3A; (ii) increases expression of H3K4me3, H3H9ac, H3K27ac; and (iii) does not modulate the expression of DNMT1, hSET1, LSD1, MBD1, p300, HDAC2, and JMJD3 epigenetic machinery proteins in an epithelial and stromal cell specific manner. In this study, we report for the first time that inhibition of PGE2-EP2/EP4 signaling modulates DNA methylation, H3 histone methylation and acetylation, and epigenetic memory machinery proteins in human endometriotic epithelial cells and stromal cells. Thus, targeting EP2 and EP4 receptors may emerge as long-term nonsteroidal therapy for treatment of active endometriotic lesions in women.


Assuntos
Compostos de Bifenilo/farmacologia , Metilação de DNA/efeitos dos fármacos , Endometriose/genética , Histonas/metabolismo , Antagonistas de Prostaglandina/farmacologia , Xantonas/farmacologia , Acetilação/efeitos dos fármacos , Linhagem Celular , Endometriose/tratamento farmacológico , Endometriose/metabolismo , Epigênese Genética/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
7.
J Ovarian Res ; 7: 17, 2014 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-24502583

RESUMO

BACKGROUND: Endometriosis is characterized by the presence of functional endometrial tissue outside of the uterine cavity. It affects 1 in 10 women of reproductive age. This chronic condition commonly leads to consequences such as pelvic pain, dysmenorrhea, infertility and an elevated risk of epithelial ovarian cancer. Despite the prevalence of endometriosis and its impact on women's lives, there are relatively few in vitro and in vivo models available for studying the complex disease biology, pathophysiology, and for use in the preclinical development of novel therapies. The goal of this study was to develop a novel three-dimensional (3D) cell culture model of ovarian endometriosis and to test whether it is more reflective of endometriosis biology than traditional two dimensional (2D) monolayer cultures. METHODS: A novel ovarian endometriosis epithelial cell line (EEC16) was isolated from a 34-year old female with severe endometriosis. After characterization of cells using in vitro assays, western blotting and RNA-sequencing, this cell line and a second, already well characterized endometriosis cell line, EEC12Z, were established as in vitro 3D spheroid models. We compared biological features of 3D spheroids to 2D cultures and human endometriosis lesions using immunohistochemistry and real-time semi-quantitative PCR. RESULTS: In comparison to normal ovarian epithelial cells, EEC16 displayed features of neoplastic transformation in in vitro assays. When cultured in 3D, EEC16 and EEC12Z showed differential expression of endometriosis-associated genes compared to 2D monolayer cultures, and more closely mimicked the molecular and histological features of human endometriosis lesions. CONCLUSIONS: To our knowledge, this represents the first report of an in vitro spheroid model of endometriosis. 3D endometriosis models represent valuable experimental tools for studying EEC biology and the development of novel therapeutic approaches.


Assuntos
Endometriose/patologia , Células Epiteliais/patologia , Doenças Ovarianas/patologia , Adulto , Biomarcadores/metabolismo , Linhagem Celular , Proliferação de Células , Endometriose/genética , Endometriose/metabolismo , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica , Genótipo , Humanos , Doenças Ovarianas/genética , Doenças Ovarianas/metabolismo , Fenótipo , Índice de Gravidade de Doença , Esferoides Celulares , Fatores de Tempo
8.
Angiogenesis ; 16(1): 223-33, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23096928

RESUMO

Cathepsin B has been shown to be important in angiogenesis; therefore, understanding its regulation in endothelial cells should provide fundamental information that will aid in the development of new treatment options. Peroxisome proliferator-activated receptors (PPARs) have been shown to have anti-inflammatory, anti-angiogenic and anti-tumorigenic properties. We explored the influence of a PPARα agonist on cathepsin B expression in human endothelial cells. The PPARα agonist, Wy14643, was found to inhibit cathepsin B protein expression. Further studies demonstrated the Wy14643-dependent but PPARα-independent suppression of cathepsin B. This has been previously described for other PPAR agonists. Wy14643 suppressed the accumulation of cathepsin B mRNA, which was accompanied by the selective suppression of a 5'-alternative splice variant. Consistent with these results, luciferase promoter assays and electrophoretic mobility shift analysis demonstrated that the suppression was facilitated by reduced binding of the transcription factors USF1/2 to an E-box within the cathepsin B promoter. Additionally, Wy14643 treatment resulted in a reduction in cathepsin B half-life, suggesting a posttranslational regulatory mechanism. Overall, our results suggest that the PPARα-dependent anti-angiogenic action of Wy14643 seems to be mediated, in part, by Wy14643-dependent but PPARα-independent regulation of cathepsin B expression.


Assuntos
Catepsina B/genética , Catepsina B/metabolismo , Células Endoteliais da Veia Umbilical Humana/enzimologia , PPAR alfa/agonistas , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Pirimidinas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/genética , Pareamento de Bases/genética , Western Blotting , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Elementos E-Box/genética , Éxons/genética , Meia-Vida , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , PPAR alfa/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Processamento de Proteína Pós-Traducional/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/enzimologia , Fatores Estimuladores Upstream/metabolismo
9.
Fertil Steril ; 99(3): 871-881.e1, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23206733

RESUMO

OBJECTIVE: To study the function of syndecan-1 (SDC1) and its potential regulator miR-10b in endometriosis. DESIGN: Experimental laboratory study. SETTING: University medical center. PATIENT(S): Not applicable. INTERVENTION(S): The human endometriotic cell line 12Z was transiently transfected with SDC1 small interfering RNA or miR-10b precursors and investigated for changes in cell behavior and gene expression. 12Z and primary eutopic endometrial stroma cells of two American Society for Reproductive Medicine class III endometriosis patients were transfected with miR-10b precursors to investigate posttranscriptional regulation of SDC1. MAIN OUTCOME MEASURE(S): Quantitative polymerase chain reaction, Western blotting, flow cytometry, 3' untranslated region luciferase assays, and zymography were used to measure miR-10b-dependent targeting of SDC1 and SDC1-dependent expression changes of proteases and interleukin-6. Altered cell behavior was monitored by Matrigel invasion assays, cell viability assays, and mitogen-activated protein kinase activation blots. RESULT(S): Knockdown of SDC1 inhibited Matrigel invasiveness by >60% but did not affect cell viability. Interleukin-6 secretion, matrix metalloproteinase-9 expression, and matrix metalloproteinase-2 activity were reduced, whereas plasminogen activator inhibitor-1 protein expression was up-regulated. miR-10b overexpression significantly down-regulated SDC1, reduced Matrigel invasiveness by 20% and cell viability by 14%, and decreased mitogen-activated protein kinase activation in response to hepatocyte growth factor. CONCLUSION(S): Syndecan-1, a target of miR-10b, inhibits epithelial endometriotic cell invasiveness through down-regulation of metalloproteinase activity and interleukin-6.


Assuntos
Endometriose/genética , Endométrio/citologia , Interleucina-6/metabolismo , MicroRNAs/genética , Sindecana-1/genética , Materiais Biocompatíveis , Linhagem Celular , Sobrevivência Celular/fisiologia , Colágeno , Combinação de Medicamentos , Endometriose/metabolismo , Endometriose/patologia , Feminino , Humanos , Laminina , Sistema de Sinalização das MAP Quinases/fisiologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Proteoglicanas , RNA Interferente Pequeno/genética , Sindecana-1/metabolismo , Transfecção
10.
Exp Dermatol ; 21(10): 751-7, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23078396

RESUMO

Peroxisome proliferator-activated receptor (PPAR) delta agonists are known to have distinct anti-inflammatory and antitumor effects; though, the knowledge regarding their mode of action has thus far been limited. Different cathepsins have been shown to be upregulated in a broad range of pathological events, such as rheumatoid arthritis, psoriasis, atherosclerosis and diverse tumor entities, for example melanoma. Recent work demonstrated that cathepsin B in particular is an important pro-angiogenic protease in various pathological conditions. We therefore analysed whether cathepsins are a valid target for PPARδ agonists. This study reveals an inhibitory effect of two commonly used PPARδ agonists, GW501516 and L-165,041, on the protein expression and enzyme activity of cathepsin B in human endothelial cells. In contrast, no inhibitory effects were observed on cathepsin L and cathepsin D protein expression after treatment with PPARδ agonists. Furthermore, the results substantiate that PPARδ activators mediate their inhibitory action in a PPARδ-dependent manner and that the underlying regulatory mechanism is not based on a transcriptional but rather on a posttranslational mode of action, via the reduction in the cathepsin B protein half-life. Mechanisms conveying the suppressive effect by 5'-alternative splicing, a 3'-UTR-dependent way or by miRNA could be excluded. The data of this study explore cathepsin B as a new valid target for PPARδ agonists in endothelial cells. The results bolster other studies demonstrating PPARδ agonists as anti-inflammatory and anticarcinogenic agents and thus might have the potential to help to develop new pharmaceutical drugs.


Assuntos
Catepsina B/antagonistas & inibidores , PPAR delta/agonistas , Regiões 3' não Traduzidas , Sequência de Bases , Catepsina B/genética , Catepsina B/metabolismo , RNA Helicases DEAD-box/antagonistas & inibidores , RNA Helicases DEAD-box/genética , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Ligantes , MicroRNAs/genética , MicroRNAs/metabolismo , PPAR delta/metabolismo , Fenoxiacetatos/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , RNA Interferente Pequeno/genética , Ribonuclease III/antagonistas & inibidores , Ribonuclease III/genética , Tiazóis/farmacologia
11.
Mol Cell Endocrinol ; 332(1-2): 306-13, 2011 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-21111772

RESUMO

Prostaglandin E2 (PGE2) plays an important role in the pathogenesis of endometriosis. We recently reported that inhibition of COX-2 decreased migration as well as invasion of human endometriotic epithelial and stromal cells. Results of the present study indicates that selective inhibition of PGE2 receptors EP2 and EP4 suppresses expression and/or activity of MMP1, MMP2, MMP3, MMP7 and MMP9 proteins and increases expression of TIMP1, TIMP2, TIMP3, and TIMP4 proteins and thereby decreases migration and invasion of human immortalized endometriotic epithelial and stromal cells into matrigel. The interactions between EP2/EP4 and MMPs are mediated through Src and ß-arrestin 1 protein complex involving MT1-MMP and EMMPRIN in human endometriotic cells. These novel findings provide an important molecular and cellular framework for further evaluation of selective inhibition of EP2 and EP4 as potential nonsteroidal therapy for endometriosis in childbearing-age women.


Assuntos
Endométrio/citologia , Células Epiteliais/patologia , Inibidores de Metaloproteinases de Matriz , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Células Estromais/patologia , Movimento Celular/fisiologia , Células Cultivadas , Dinoprostona/metabolismo , Endometriose/metabolismo , Endometriose/patologia , Células Epiteliais/metabolismo , Feminino , Humanos , RNA Interferente Pequeno/metabolismo , Receptores de Prostaglandina E Subtipo EP2/genética , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Células Estromais/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo
12.
Fertil Steril ; 93(8): 2498-506, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20207353

RESUMO

OBJECTIVE: To determine interactions between prostaglandin (PG) E(2) signaling and proliferation of endometriotic cells to increase our knowledge about PGE(2) signaling in the pathogenesis of endometriosis in humans. DESIGN: Immortalized human endometriotic epithelial and stromal cells were used as an in vitro model. Effects of inhibition of PGE(2) receptors on proliferation of endometriotic cells and associated cell cycle regulation were determined. SETTING: College Veterinary Medicine and Biomedical Sciences, Texas A&M University. PATIENT(S): Not available. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Cell proliferation, cell viability, cell cycle, regulation of cyclins, cyclin-dependent kinases, and cyclin-dependent kinase inhibitors. RESULT(S): Selective blockade of EP2/EP4 inhibited proliferation of human endometriotic cells by inducing cell cycle arrest at the G(1)-S and G(2)-M checkpoints in epithelial cells and at the G(2)-M checkpoint in stromal cells. This cell cycle arrest during specific checkpoints was associated with distinct regulation of respective cyclins and cyclin-dependent kinases. Inhibition of EP1 did not decrease endometriotic cell proliferation. CONCLUSION(S): For the first time data from the present study provide a direct molecular link between PGE(2) signaling and proliferation of endometriotic cells and suggest that inhibition of EP2/EP4 could be a potential nonestrogen (E) treatment option for endometriosis in women.


Assuntos
Endometriose/metabolismo , Receptores de Prostaglandina E/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Compostos de Bifenilo/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/antagonistas & inibidores , Endometriose/tratamento farmacológico , Endométrio/citologia , Endométrio/efeitos dos fármacos , Feminino , Humanos , Antagonistas de Prostaglandina/farmacologia , RNA Interferente Pequeno/farmacologia , Receptores de Prostaglandina E Subtipo EP2 , Receptores de Prostaglandina E Subtipo EP4 , Células Estromais/metabolismo , Xantonas/farmacologia
13.
Gynecol Obstet Invest ; 70(1): 23-33, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20130413

RESUMO

OBJECTIVE(S): To determine whether nuclear factor-kappaB (NF-kappaB) is constitutively and tumor necrosis factor (TNF)-dependently activated in endometriotic cells, whether trichostatin A (TSA) can suppress NF-kappaB activation and suppress TRAF2/6 and TAK1, and whether TSA and caffeic acid phenyl ester can suppress constitutive and H(2)O(2)-stimulated proliferation of endometriotic cells. METHODS: Two endometriotic cell lines and an endometrial stromal cell line were used as an in vitro model. Electrophoretic mobility shift analysis was used to determine NF-kappaB activation and possible suppression by TSA. Western blot analysis was used to determine whether TSA suppresses phosphorylation of IkappaBalpha, phosphorylation of p65 in the cytoplasm and nuclear translocation, and the expression of TRAF2/6 and TAK1. RESULTS: NF-kappaB was constitutively activated in endometriotic cells, but only minimally in endometrial cells. TNFalpha stimulation activated NF-kappaB through induction of IkappaB phosphorylation, but the activation can be suppressed by TSA. TSA also attenuated constitutive and TNF-dependent p65 phosphorylation and nuclear translocation in endometriotic cells. TRAF2, TRAF6 and TAK1 were constitutively activated and were unaffected by TSA treatment. CONCLUSIONS: NF-kappaB activation may play a critical role in the pathogenesis in endometriosis. Targeting NF-kappaB with histone deacetylase inhibitors or other compounds might hold promise as novel therapeutics for endometriosis.


Assuntos
Endométrio/citologia , Ácidos Hidroxâmicos/farmacologia , Células Estromais/efeitos dos fármacos , Fator 2 Associado a Receptor de TNF/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Fator de Transcrição RelA/metabolismo , Ácidos Cafeicos/farmacologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular Transformada , Endometriose/tratamento farmacológico , Endometriose/metabolismo , Endometriose/patologia , Feminino , Inibidores de Histona Desacetilases/farmacologia , Humanos , Peróxido de Hidrogênio/farmacologia , Quinase I-kappa B/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Oxidantes/farmacologia , Álcool Feniletílico/análogos & derivados , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Células Estromais/citologia , Células Estromais/metabolismo
14.
Clin Cancer Res ; 15(21): 6541-50, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19861460

RESUMO

PURPOSE: Glioblastoma spheroid cultures are enriched in tumor stem-like cells and therefore may be more representative of the respective primary tumors than conventional monolayer cultures. We exploited the glioma spheroid culture model to find novel tumor-relevant genes. EXPERIMENTAL DESIGN: We carried out array-based comparative genomic hybridization of spheroid cultures derived from 20 glioblastomas. Microarray-based gene expression analysis was applied to determine genes with differential expression compared with normal brain tissue and to nonneoplastic brain spheroids in glioma spheroid cultures. The protein expression levels of three candidates were determined by immunohistochemistry on tissue microarrays and correlated with clinical outcome. Functional analysis of PDPN was done. RESULTS: Genomic changes in spheroid cultures closely resembled those detected in primary tumors of the corresponding patients. In contrast, genomic changes in serum-grown monolayer cultures established from the same patients did not match well with the respective primary tumors. Microarray-based gene expression analysis of glioblastoma spheroid cultures identified a set of novel candidate genes being upregulated or downregulated relative to normal brain. Quantitative real-time PCR analyses of 8 selected candidate genes in 20 clinical glioblastoma samples validated the microarray findings. Immunohistochemistry on tissue microarrays revealed that expression of AJAP1, EMP3, and PDPN was significantly associated with overall survival of astrocytic glioma patients. Invasive capacity and RhoA activity were decreased in PDPN-silenced spheroids. CONCLUSION: We identified a set of novel candidate genes that likely play a role in glioblastoma pathogenesis and implicate AJAP1, EMP3, and PDPN as molecular markers associated with the clinical outcome of glioma patients.


Assuntos
Neoplasias Encefálicas/genética , Perfilação da Expressão Gênica , Glioblastoma/genética , Células-Tronco Neoplásicas , Esferoides Celulares , Biomarcadores Tumorais , Encéfalo/metabolismo , Técnicas de Cultura de Células , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Esferoides Celulares/metabolismo , Células Tumorais Cultivadas
15.
Mol Biol Cell ; 20(15): 3598-607, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19515834

RESUMO

Gain- and loss-of-function studies indicate that the adherens junction protein shrew-1 acts as a novel modulator of E-cadherin internalization induced by epithelial growth factor (EGF) or E-cadherin function-blocking antibody during epithelial cell dynamics. Knocking down shrew-1 in MCF-7 carcinoma cells preserves E-cadherin surface levels upon EGF stimulation. Overexpression of shrew-1 leads to preformation of an E-cadherin/EGF receptor (EGFR) HER2/src-kinase/shrew-1 signaling complex and accelerated E-cadherin internalization. Shrew-1 is not sufficient to stimulate E-cadherin internalization, but facilitates the actions of EGFR and thus may promote malignant progression in breast cancer cells with constitutive EGFR stimulation by reducing surface E-cadherin expression.


Assuntos
Junções Aderentes/efeitos dos fármacos , Caderinas/metabolismo , Moléculas de Adesão Celular/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Junções Aderentes/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Receptores ErbB/metabolismo , Imunofluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Immunoblotting , Glândulas Mamárias Humanas/metabolismo , Microscopia Confocal , Interferência de RNA , Receptor ErbB-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/metabolismo
16.
Mol Endocrinol ; 23(8): 1291-305, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19407222

RESUMO

Endometriosis is a benign chronic gynecological disease of reproductive-age women characterized by the presence of functional endometrial tissues outside the uterine cavity. It is an estrogen-dependent disease. Current treatment modalities to inhibit biosynthesis and actions of estrogen compromise menstruation, pregnancy, and the reproductive health of women and fail to prevent reoccurrence of disease. There is a critical need to identify new specific signaling modules for non-estrogen-targeted therapies for endometriosis. In our previous study, we reported that selective inhibition of cyclooxygenase-2 prevented survival, migration, and invasion of human endometriotic epithelial and stromal cells, which was due to decreased prostaglandin E(2) (PGE(2)) production. In this study, we determined mechanisms through which PGE(2) promoted survival of human endometriotic cells. Results of the present study indicate that 1) PGE(2) promotes survival of human endometriotic cells through EP2 and EP4 receptors by activating ERK1/2, AKT, nuclear factor-kappaB, and beta-catenin signaling pathways; 2) selective inhibition of EP2 and EP4 suppresses these cell survival pathways and augments interactions between proapoptotic proteins (Bax and Bad) and antiapoptotic proteins (Bcl-2/Bcl-XL), facilitates the release of cytochrome c, and thus activates caspase-3/poly (ADP-ribose) polymerase-mediated intrinsic apoptotic pathways; and 3) these PGE(2) signaling components are more abundantly expressed in ectopic endometriosis tissues compared with eutopic endometrial tissues during the menstrual cycle in women. These novel findings may provide an important molecular framework for further evaluation of selective inhibition of EP2 and EP4 as potential therapy, including nonestrogen target, to expand the spectrum of currently available treatment options for endometriosis in women.


Assuntos
Apoptose , Endométrio/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Prostaglandina E/antagonistas & inibidores , beta Catenina/metabolismo , Endometriose/metabolismo , Endometriose/patologia , Endométrio/patologia , Feminino , Humanos , Microscopia de Fluorescência/métodos , Mitocôndrias/patologia , Modelos Biológicos , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP2 , Receptores de Prostaglandina E Subtipo EP4
17.
Genes Chromosomes Cancer ; 48(3): 229-38, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19025795

RESUMO

Nonresectable ependymomas are associated with poor prognosis despite intensive radiochemotherapy and radiation. The molecular pathogenesis of ependymoma initiation and progression is largely unknown. We here present a case of therapy-refractory, progressive ependymoma with cerebrospinal as well as extraneural metastases, which allowed us for the first time to follow the stepwise accumulation of chromosome aberrations during disease progression. Genome-wide DNA copy-number analysis showed sequential deletions on chromosomes 1, 9, and 14 as well as a homozygous deletion of the CDKN2A locus, underscoring its role in tumor progression. Gradual loss at 1p36 was associated with loss of protein expression of the putative tumor suppressor gene AJAP1/SHREW1. In summary, this is the first report on acquired genomic aberrations in ependymoma over time pointing to novel candidate tumor suppressor genes. This analysis provides molecular insights into the chronology of genetic events in this case from initial localized tumor to widespread metastasized disease.


Assuntos
Neoplasias Encefálicas/genética , Deleção Cromossômica , Ependimoma/genética , Adolescente , Neoplasias Encefálicas/patologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 14/genética , Cromossomos Humanos Par 9/genética , Ependimoma/patologia , Ependimoma/secundário , Evolução Fatal , Feminino , Expressão Gênica , Genes Supressores de Tumor , Humanos , Análise em Microsséries , Neoplasias Peritoneais/patologia , Neoplasias Peritoneais/secundário , Neoplasias da Coluna Vertebral/patologia , Neoplasias da Coluna Vertebral/secundário , Células Tumorais Cultivadas
18.
Fertil Steril ; 91(5 Suppl): 2199-209, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18722604

RESUMO

OBJECTIVE: To determine whether a mixed population of immortalized human endometriosis epithelial and stromal cells is able to induce peritoneal endometriosis in nude mice. DESIGN: Prospective experimental study. Human immortalized endometriosis epithelial and stromal cells were xenografted into ovariectomized nude mice. Macroscopically, the number of induced endometriosis-like lesions and their color were determined. Microscopically, histomorphology of endometriosis glands and their structure were analyzed, and comparisons were made with tissue from spontaneous endometriosis in women. SETTING: College of Veterinary Medicine and Biomedical Sciences, Texas A&M University. ANIMALS: Seven ovariectomized nude mice. INTERVENTION(S): Minimal invasive procedures were performed to administer estrogen pellets and transplant immortalized human endometriosis epithelial and stromal cells into nude mice. MAIN OUTCOME MEASURE(S): Peritoneal endometriosis-like lesions induced in nude mice were characterized and compared with spontaneous peritoneal endometriosis in women. RESULT(S): Xenografts of human immortalized endometriosis epithelial and stromal cells into the peritoneal cavity of the recipient nude mice are able to proliferate, attach, invade, reorganize, and establish peritoneal endometriosis. Endometriosis glands at different stages of growth were present in induced endometriosis-like lesions. Proliferating cell nuclear antigen, metalloproteinase 2, estrogen receptor-alpha, cyclooxygenase-2, and prostaglandin E(2) receptors EP2 and EP4 proteins were expressed in both endometriosis glandular epithelial and stromal cells of the induced endometriosis-like lesions. CONCLUSION(S): This xenograft model could be used as a potential experimental tool to understand the molecular and cellular aspects of the pathogenesis of endometriosis in humans.


Assuntos
Endometriose/patologia , Células Epiteliais/transplante , Doenças Peritoneais/patologia , Células Estromais/patologia , Células Estromais/transplante , Animais , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Humanos , Camundongos , Camundongos Nus , Ovariectomia , Especificidade da Espécie , Transplante Heterólogo
19.
PLoS One ; 3(10): e3510, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18946507

RESUMO

Exported proteases of Helicobacter pylori (H. pylori) are potentially involved in pathogen-associated disorders leading to gastric inflammation and neoplasia. By comprehensive sequence screening of the H. pylori proteome for predicted secreted proteases, we retrieved several candidate genes. We detected caseinolytic activities of several such proteases, which are released independently from the H. pylori type IV secretion system encoded by the cag pathogenicity island (cagPAI). Among these, we found the predicted serine protease HtrA (Hp1019), which was previously identified in the bacterial secretome of H. pylori. Importantly, we further found that the H. pylori genes hp1018 and hp1019 represent a single gene likely coding for an exported protein. Here, we directly verified proteolytic activity of HtrA in vitro and identified the HtrA protease in zymograms by mass spectrometry. Overexpressed and purified HtrA exhibited pronounced proteolytic activity, which is inactivated after mutation of Ser205 to alanine in the predicted active center of HtrA. These data demonstrate that H. pylori secretes HtrA as an active protease, which might represent a novel candidate target for therapeutic intervention strategies.


Assuntos
Helicobacter pylori/enzimologia , Peptídeo Hidrolases/metabolismo , Processamento de Proteína Pós-Traducional , Serina Endopeptidases/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Caseínas/metabolismo , Biologia Computacional , Espaço Extracelular/enzimologia , Espaço Extracelular/metabolismo , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Helicobacter pylori/metabolismo , Humanos , Dados de Sequência Molecular , Sinais Direcionadores de Proteínas/genética , Proteoma/análise , Análise de Sequência de DNA , Serina Endopeptidases/genética
20.
PLoS One ; 3(7): e2767, 2008 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-18648515

RESUMO

Targeting signals direct proteins to their extra- or intracellular destination such as the plasma membrane or cellular organelles. Here we investigated the structure and function of exceptionally long signal peptides encompassing at least 40 amino acid residues. We discovered a two-domain organization ("NtraC model") in many long signals from vertebrate precursor proteins. Accordingly, long signal peptides may contain an N-terminal domain (N-domain) and a C-terminal domain (C-domain) with different signal or targeting capabilities, separable by a presumably turn-rich transition area (tra). Individual domain functions were probed by cellular targeting experiments with fusion proteins containing parts of the long signal peptide of human membrane protein shrew-1 and secreted alkaline phosphatase as a reporter protein. As predicted, the N-domain of the fusion protein alone was shown to act as a mitochondrial targeting signal, whereas the C-domain alone functions as an export signal. Selective disruption of the transition area in the signal peptide impairs the export efficiency of the reporter protein. Altogether, the results of cellular targeting studies provide a proof-of-principle for our NtraC model and highlight the particular functional importance of the predicted transition area, which critically affects the rate of protein export. In conclusion, the NtraC approach enables the systematic detection and prediction of cryptic targeting signals present in one coherent sequence, and provides a structurally motivated basis for decoding the functional complexity of long protein targeting signals.


Assuntos
Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Fosfatase Alcalina/metabolismo , Aminoácidos/química , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Clonagem Molecular , Densitometria , Humanos , Mitocôndrias/metabolismo , Modelos Biológicos , Oligonucleotídeos/química , Peptídeos/química , Sinais Direcionadores de Proteínas , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA