Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell Rep Methods ; 4(4): 100740, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38521059

RESUMO

Intracellular signaling plays essential roles in various cell types. In the central nervous system, signaling cascades are strictly regulated in a spatiotemporally specific manner to govern brain function; for example, presynaptic cyclic adenosine monophosphate (cAMP) can enhance the probability of neurotransmitter release. In the last decade, channelrhodopsin-2 has been engineered for subcellular targeting using localization tags, but optogenetic tools for intracellular signaling are not well developed. Therefore, we engineered a selective presynaptic fusion tag for photoactivated adenylyl cyclase (bPAC-Syn1a) and found its high localization at presynaptic terminals. Furthermore, an all-optical electrophysiological method revealed rapid and robust short-term potentiation by bPAC-Syn1a at brain stem-amygdala synapses in acute brain slices. Additionally, bPAC-Syn1a modulated mouse immobility behavior. These results indicate that bPAC-Syn1a can manipulate presynaptic cAMP signaling in vitro and in vivo. The all-optical manipulation technique developed in this study can help further elucidate the dynamic regulation of various cellular functions.


Assuntos
Adenilil Ciclases , AMP Cíclico , Plasticidade Neuronal , Terminações Pré-Sinápticas , Animais , Masculino , Camundongos , Adenilil Ciclases/metabolismo , Adenilil Ciclases/genética , AMP Cíclico/metabolismo , Células HEK293 , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/fisiologia , Optogenética/métodos , Terminações Pré-Sinápticas/metabolismo , Ratos
2.
Nat Commun ; 13(1): 7913, 2022 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-36585411

RESUMO

Feeding behavior is adaptively regulated by external and internal environment, such that feeding is suppressed when animals experience pain, sickness, or fear. While the lateral parabrachial nucleus (lPB) plays key roles in nociception and stress, neuronal pathways involved in feeding suppression induced by fear are not fully explored. Here, we investigate the parasubthalamic nucleus (PSTN), located in the lateral hypothalamus and critically involved in feeding behaviors, as a target of lPB projection neurons. Optogenetic activation of lPB-PSTN terminals in male mice promote avoidance behaviors, aversive learning, and suppressed feeding. Inactivation of the PSTN and lPB-PSTN pathway reduces fear-induced feeding suppression. Activation of PSTN neurons expressing pituitary adenylate cyclase-activating polypeptide (PACAP), a neuropeptide enriched in the PSTN, is sufficient for inducing avoidance behaviors and feeding suppression. Blockade of PACAP receptors impaires aversive learning induced by lPB-PSTN photomanipulation. These findings indicate that lPB-PSTN pathway plays a pivotal role in fear-induced feeding suppression.


Assuntos
Núcleos Parabraquiais , Camundongos , Masculino , Animais , Núcleos Parabraquiais/metabolismo , Medo , Dor , Região Hipotalâmica Lateral/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo
3.
Front Surg ; 9: 864255, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35647017

RESUMO

An ectopic parathyroid adenoma (EPA) is a rare entity. The aim of this study was to report our experience in the preoperative localization and surgical management of EPAs. This was a multicenter retrospective study involving patients diagnosed with an EPA (three males and seven females) from January 2005 to November 2021. The clinical features, preoperative management, and surgical procedures were analyzed. A cervical neck ultrasound was performed in all patients and showed a focus in eight patients. Cervicothoracic enhanced computed tomography was performed in all patients and showed a focus in nine patients. The 99mTc-MIBI scintigraphy was performed in eight patients and showed uptake in six of them. We performed a neck dissection and thoracotomy in one patient, a thoracoscopy in one patient, surgery with a focused approach in seven patients, four of whom were injected with indigo carmine blue, and surgery with a bilateral approach in one patient. 1 h following the parathyroidectomy, the parathyroid hormone (PTH) concentration was decreased to 40-80% of the baseline value. Establishing a preoperative diagnosis of an EPA is challenging for the surgeon, despite the progress in the morphologic assessment. An intraoperative PTH assay and injection of indigo carmine have been shown to be valuable tools in the appropriate surgical management of an EPA.

4.
Nat Commun ; 12(1): 3386, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099644

RESUMO

During early pregnancy in the mouse, nidatory estrogen (E2) stimulates endometrial receptivity by activating a network of signaling pathways that is not yet fully characterized. Here, we report that bone morphogenetic proteins (BMPs) control endometrial receptivity via a conserved activin receptor type 2 A (ACVR2A) and SMAD1/5 signaling pathway. Mice were generated to contain single or double conditional deletion of SMAD1/5 and ACVR2A/ACVR2B receptors using progesterone receptor (PR)-cre. Female mice with SMAD1/5 deletion display endometrial defects that result in the development of cystic endometrial glands, a hyperproliferative endometrial epithelium during the window of implantation, and impaired apicobasal transformation that prevents embryo implantation and leads to infertility. Analysis of Acvr2a-PRcre and Acvr2b-PRcre pregnant mice determined that BMP signaling occurs via ACVR2A and that ACVR2B is dispensable during embryo implantation. Therefore, BMPs signal through a conserved endometrial ACVR2A/SMAD1/5 pathway that promotes endometrial receptivity during embryo implantation.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Implantação do Embrião , Infertilidade Feminina/genética , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animais , Biópsia , Modelos Animais de Doenças , Endométrio/metabolismo , Endométrio/patologia , Estrogênios/metabolismo , Feminino , Humanos , Camundongos , Camundongos Knockout , Gravidez , Transdução de Sinais/fisiologia , Proteína Smad1/análise , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/análise , Proteína Smad5/genética , Proteína Smad5/metabolismo
5.
Growth Horm IGF Res ; 41: 34-41, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30005335

RESUMO

OBJECTIVE: Trophoblast survival is regulated by cytokines and growth factors. While the pharmacological levels (10-100 ng/mL) of tumor necrosis factor (TNF)- α affect trophoblasts survival in vitro, the effects of the physiological levels (1-10 pg/mL) of TNF-α remain unknown. We investigated the effects of the physiological levels of TNF-α on proliferation and apoptosis of human trophoblast cells by using BeWo cells. Insulin-like growth factor (IGF)-I is also a potent regulator of trophoblast survival and has been known to exert synergistic effects with other hormones. The interaction of IGF-I and TNF-α on BeWo cells survival was also examined. METHODS: After incubating BeWo under the presence of TNF-α (10-105 pg/mL) and IGF-I (102 ng/mL), we assessed cell number by WST-1 assay and cell proliferation by BrdU uptake assay and immunocytochemistry with anti-Ki67 antibody. Apoptosis was evaluated by TUNEL assay and caspase-3, 8 activity assays. RESULTS: Under the presence of IGF-I, cell number, BrdU uptake, and Ki-67 expression of BeWo were dose-dependently enhanced by low TNF-α (10-102 pg/mL), while no such effects were detected without IGF-I. Higher levels of TNF-α (104-105 pg/mL) showed inhibiting effects on cell number and cell proliferation. The number of TUNEL positive cells were decreased and caspase activities were suppressed by lower levels (10-102 pg/mL) of TNF-α and IGF-I independently. Higher levels of TNF-α (104-105 pg/mL) showed promoting effects on apoptosis irrespective of IGF-I. CONCLUSION: The physiological levels of TNF-α and IGF-I had synergetic effects on enhancing cell proliferation and also independently inhibited apoptosis of Bewo cells.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Fator de Crescimento Insulin-Like I/administração & dosagem , Trofoblastos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Células Cultivadas , Citocinas/metabolismo , Combinação de Medicamentos , Humanos , Trofoblastos/metabolismo , Trofoblastos/patologia
6.
Biol Reprod ; 93(2): 37, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26108791

RESUMO

Repeated and dramatic pregnancy-induced uterine enlargement and remodeling throughout reproductive life suggests the existence of uterine smooth muscle stem/progenitor cells. The aim of this study was to isolate and characterize stem/progenitor-like cells from human myometrium through identification of specific surface markers. We here identify CD49f and CD34 as markers to permit selection of the stem/progenitor cell-like population from human myometrium and show that human CD45(-) CD31(-) glycophorin A(-) and CD49f(+) CD34(+) myometrial cells exhibit stem cell-like properties. These include side population phenotypes, an undifferentiated status, high colony-forming ability, multilineage differentiation into smooth muscle cells, osteoblasts, adipocytes, and chondrocytes, and in vivo myometrial tissue reconstitution following xenotransplantation. Furthermore, CD45(-) CD31(-) glycophorin A(-) and CD49f(+) CD34(+) myometrial cells proliferate under hypoxic conditions in vitro and, compared with the untreated nonpregnant myometrium, show greater expansion in the estrogen-treated nonpregnant myometrium and further in the pregnant myometrium in mice upon xenotransplantation. These results suggest that the newly identified myometrial stem/progenitor-like cells influenced by hypoxia and sex steroids may participate in pregnancy-induced uterine enlargement and remodeling, providing novel insights into human myometrial physiology.


Assuntos
Antígenos CD34/genética , Antígenos CD34/fisiologia , Integrina alfa6/genética , Integrina alfa6/fisiologia , Miométrio/metabolismo , Células-Tronco/fisiologia , Útero/fisiologia , Animais , Diferenciação Celular , Hipóxia Celular , Linhagem da Célula/genética , Feminino , Glicoforinas/biossíntese , Glicoforinas/genética , Células-Tronco Hematopoéticas , Humanos , Camundongos , Miométrio/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Gravidez
7.
J Clin Invest ; 123(6): 2539-50, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23676498

RESUMO

Abnormalities in cell-cell communication and growth factor signaling pathways can lead to defects in maternal-fetal interactions during pregnancy, including immunologic rejection of the fetal/placental unit. In this study, we discovered that bone morphogenetic protein receptor type 2 (BMPR2) is essential for postimplantation physiology and fertility. Despite normal implantation and early placental/fetal development, deletion of Bmpr2 in the uterine deciduae of mice triggered midgestation abnormalities in decidualization that resulted in abnormal vascular development, trophoblast defects, and a deficiency of uterine natural killer cells. Absence of BMPR2 signaling in the uterine decidua consequently suppressed IL-15, VEGF, angiopoietin, and corin signaling. Disruption of these pathways collectively lead to placental abruption, fetal demise, and female sterility, thereby placing BMPR2 at a central point in the regulation of several physiologic signaling pathways and events at the maternal-fetal interface. Since trophoblast invasion and uterine vascular modification are implicated in normal placentation and fetal growth in humans, our findings suggest that abnormalities in uterine BMPR2-mediated signaling pathways can have catastrophic consequences in women for the maintenance of pregnancy.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Decídua/metabolismo , Implantação do Embrião , Manutenção da Gravidez , Angiopoietinas/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Diferenciação Celular , Proliferação de Células , Decídua/imunologia , Decídua/patologia , Estradiol/sangue , Feminino , Retardo do Crescimento Fetal/genética , Expressão Gênica , Células Gigantes/patologia , Infertilidade Feminina/genética , Interleucina-15/metabolismo , Células Matadoras Naturais/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placenta/irrigação sanguínea , Placenta/patologia , Gravidez , Progesterona/sangue , Serina Endopeptidases/metabolismo , Transdução de Sinais , Trofoblastos/patologia , Útero/imunologia , Útero/metabolismo , Útero/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
PLoS Genet ; 7(10): e1002320, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22028666

RESUMO

The transforming growth factor ß (TGFß) superfamily proteins are principle regulators of numerous biological functions. Although recent studies have gained tremendous insights into this growth factor family in female reproduction, the functions of the receptors in vivo remain poorly defined. TGFß type 1 receptor (TGFBR1), also known as activin receptor-like kinase 5, is the major type 1 receptor for TGFß ligands. Tgfbr1 null mice die embryonically, precluding functional characterization of TGFBR1 postnatally. To study TGFBR1-mediated signaling in female reproduction, we generated a mouse model with conditional knockout (cKO) of Tgfbr1 in the female reproductive tract using anti-Müllerian hormone receptor type 2 promoter-driven Cre recombinase. We found that Tgfbr1 cKO females are sterile. However, unlike its role in growth differentiation factor 9 (GDF9) signaling in vitro, TGFBR1 seems to be dispensable for GDF9 signaling in vivo. Strikingly, we discovered that the Tgfbr1 cKO females develop oviductal diverticula, which impair embryo development and transit of embryos to the uterus. Molecular analysis further demonstrated the dysregulation of several cell differentiation and migration genes (e.g., Krt12, Ace2, and MyoR) that are potentially associated with female reproductive tract development. Moreover, defective smooth muscle development was also revealed in the uteri of the Tgfbr1 cKO mice. Thus, TGFBR1 is required for female reproductive tract integrity and function, and disruption of TGFBR1-mediated signaling leads to catastrophic structural and functional consequences in the oviduct and uterus.


Assuntos
Desenvolvimento Embrionário/genética , Fator 9 de Diferenciação de Crescimento/metabolismo , Músculo Liso/crescimento & desenvolvimento , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fenômenos Reprodutivos Fisiológicos/genética , Útero/embriologia , Animais , Células Cultivadas , Divertículo/genética , Divertículo/patologia , Tubas Uterinas/metabolismo , Tubas Uterinas/patologia , Feminino , Fertilidade/genética , Fator 9 de Diferenciação de Crescimento/genética , Células HEK293 , Humanos , Integrases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso/metabolismo , Progesterona/sangue , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Útero/anormalidades
9.
Mol Endocrinol ; 25(10): 1740-59, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21868453

RESUMO

Connective tissue growth factor (CTGF) is a cysteine-rich protein the synthesis and secretion of which are hypothesized to be selectively regulated by activins and other members of the TGF-ß superfamily. To investigate the in vivo roles of CTGF in female reproduction, we generated Ctgf ovarian and uterine conditional knockout (cKO) mice. Ctgf cKO mice exhibit severe subfertility and multiple reproductive defects including disrupted follicle development, decreased ovulation rates, increased numbers of corpus luteum, and smaller but functionally normal uterine horns. Steroidogenesis is disrupted in the Ctgf cKO mice, leading to increased levels of serum progesterone. We show that disrupted follicle development is accompanied by a significant increase in granulosa cell apoptosis. Moreover, despite normal cumulus expansion, Ctgf cKO mice exhibit a significant decrease in oocytes ovulated, likely due to impaired ovulatory process. During analyses of mRNA expression, we discovered that Ctgf cKO granulosa cells show gene expression changes similar to our previously reported granulosa cell-specific knockouts of activin and Smad4, the common TGF-ß family intracellular signaling protein. We also discovered a significant down-regulation of Adamts1, a progesterone-regulated gene that is critical for the remodeling of extracellular matrix surrounding granulosa cells of preovulatory follicles. These findings demonstrate that CTGF is a downstream mediator in TGF-ß and progesterone signaling cascades and is necessary for normal follicle development and ovulation.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Folículo Ovariano/crescimento & desenvolvimento , Ovulação/fisiologia , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS1 , Animais , Apoptose , Fator de Crescimento do Tecido Conjuntivo/deficiência , Fator de Crescimento do Tecido Conjuntivo/genética , Corpo Lúteo/crescimento & desenvolvimento , Corpo Lúteo/metabolismo , Corpo Lúteo/patologia , Células do Cúmulo/metabolismo , Células do Cúmulo/patologia , Decídua/metabolismo , Decídua/patologia , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Infertilidade Feminina/sangue , Infertilidade Feminina/metabolismo , Infertilidade Feminina/patologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Folículo Ovariano/metabolismo , Folículo Ovariano/patologia , Progesterona/sangue , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Recombinação Genética/genética , Fatores de Tempo
10.
PLoS One ; 5(4): e10387, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20442847

RESUMO

BACKGROUND: The human endometrium undergoes cyclical regeneration throughout a woman's reproductive life. Ectopic implantation of endometrial cells through retrograde menstruation gives rise to endometriotic lesions which affect approximately 10% of reproductive-aged women. The high regenerative capacity of the human endometrium at eutopic and ectopic sites suggests the existence of stem/progenitor cells and a unique angiogenic system. The objective of this study was to isolate and characterize putative endometrial stem/progenitor cells and to address how they might be involved in the physiology of endometrium. METHODOLOGY/PRINCIPAL FINDINGS: We found that approximately 2% of the total cells obtained from human endometrium displayed a side population (SP) phenotype, as determined by flow cytometric analysis of Hoechst-stained cells. The endometrial SP (ESP) cells exhibited preferential expression of several endothelial cell markers compared to endometrial main population (EMP) cells. A medium specific for endothelial cell culture enabled ESP cells to proliferate and differentiate into various types of endometrial cells, including glandular epithelial, stromal and endothelial cells in vitro, whereas in the same medium, EMP cells differentiated only into stromal cells. Furthermore, ESP cells, but not EMP cells, reconstituted organized endometrial tissue with well-delineated glandular structures when transplanted under the kidney capsule of severely immunodeficient mice. Notably, ESP cells generated endothelial cells that migrated into the mouse kidney parenchyma and formed mature blood vessels. This potential for in vivo angiogenesis and endometrial cell regeneration was more prominent in the ESP fraction than in the EMP fraction, as the latter mainly gave rise to stromal cells in vivo. CONCLUSIONS/SIGNIFICANCE: These results indicate that putative endometrial stem cells are highly enriched in the ESP cells. These unique characteristics suggest that ESP cells might drive physiological endometrial regeneration and be involved in the pathogenesis of endometriosis.


Assuntos
Endométrio/citologia , Neovascularização Fisiológica , Regeneração , Células-Tronco/citologia , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/crescimento & desenvolvimento , Diferenciação Celular , Proliferação de Células , Transplante de Células , Células Cultivadas , Endométrio/fisiologia , Feminino , Rim , Camundongos , Camundongos SCID
11.
J Immunol ; 182(11): 7074-84, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19454705

RESUMO

Innate mucosal immune responses, including recognition of pathogen-associated molecular patterns through Toll-like receptors, play an important role in preventing infection in the female reproductive tract (FRT). Damaged cells release nucleotides, including ATP and uridine 5'-diphosphoglucose (UDP-glucose), during inflammation and mechanical stress. We show in this report that P2RY14, a membrane receptor for UDP-glucose, is exclusively expressed in the epithelium, but not the stroma, of the FRT in humans and mice. P2RY14 and several proinflammatory cytokines, such as IL-8, are up-regulated in the endometria of patients with pelvic inflammatory disease. UDP-glucose stimulated IL-8 production via P2RY14 in human endometrial epithelial cells but not stromal cells. Furthermore, UDP-glucose enhanced neutrophil chemotaxis in the presence of a human endometrial epithelial cell line in an IL-8-dependent manner. Administration of UDP-glucose into the mouse uterus induced expression of macrophage inflammatory protein-2 and keratinocyte-derived cytokine, two murine chemokines that are functional homologues of IL-8, and augmented endometrial neutrophil recruitment. Reduced expression of P2RY14 by small interfering RNA gene silencing attenuated LPS- or UDP-glucose-induced leukocytosis in the mouse uterus. These results suggest that UDP-glucose and its receptor P2RY14 are key front line players able to trigger innate mucosal immune responses in the FRT bypassing the recognition of pathogen-associated molecular patterns. Our findings would significantly impact the strategic design of therapies to modulate mucosal immunity by targeting P2RY14.


Assuntos
Genitália Feminina/patologia , Imunidade Inata , Interleucina-8/genética , Mucosa/imunologia , Receptores Purinérgicos P2/fisiologia , Regulação para Cima/genética , Animais , Quimiotaxia , Endométrio/metabolismo , Endométrio/patologia , Epitélio/metabolismo , Feminino , Genitália Feminina/imunologia , Humanos , Interleucina-8/biossíntese , Camundongos , Neutrófilos/fisiologia , Receptores Purinérgicos P2Y , Uridina Difosfato Glucose/farmacologia , Útero
12.
Mol Hum Reprod ; 14(1): 17-22, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18178606

RESUMO

Prolonged exposure to unopposed estrogen in the absence of progesterone gives rise to endometrial hyperplasia and carcinoma. Post-ovulatory progesterone is necessary for the proper growth and differentiation of endometrial epithelial cells (EECs). Progesterone exposure induces the endometrial production of numerous bioactive substances, one of which is the glycoprotein, glycodelin (Gd). We investigated the role of Gd in cell cycle progression and cell growth to better understand how Gd affects EEC behavior and endometrial cancer pathogenesis. Ishikawa cells, a well-differentiated human endometrial epithelial cancer cell line, were transfected with expression plasmids encoding enhanced green fluorescent protein (EGFP) or EGFP-fused Gd (EGFP-Gd). They were then subjected to a cell proliferation assay, flow cytometry cell cycle analysis and RT-PCR analysis of cyclin-dependent kinase inhibitors (CDKIs) including p21, p27 and p16. Overexpression of EGFP-Gd resulted in a reduction of cell proliferation activity, an accumulation of G1-phase cells and up-regulation of p21, p27 and p16 mRNAs. Furthermore, progesterone-induced inhibition of Ishikawa cell growth was partially attenuated by Gd knockdown using siRNA. These results indicate that Gd causes inhibition of G1/S progression together with up-regulation of CDKIs thereby reducing cell growth. Thus, progesterone-induced expression of Gd may, at least in part, contribute to the suppression of endometrial epithelial growth observed during the secretory phase.


Assuntos
Proliferação de Células/efeitos dos fármacos , Endométrio/citologia , Células Epiteliais/efeitos dos fármacos , Glicoproteínas/fisiologia , Proteínas da Gravidez/fisiologia , Progesterona/farmacologia , Fase S/efeitos dos fármacos , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Quinases Ciclina-Dependentes/antagonistas & inibidores , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Citometria de Fluxo , Glicodelina , Glicoproteínas/genética , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Immunoblotting , Imunoprecipitação , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fase S/genética , Fase S/fisiologia
13.
Endocrinology ; 149(3): 1227-34, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18063684

RESUMO

Progesterone induces decidual transformation of estrogen-primed human endometrial stromal cells (hESCs), critical for implantation and maintenance of pregnancy, through activation of many signaling pathways involving protein kinase A and signal transducer and activator of transcription (STAT)-5. We have previously shown that kinase activation of v-src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog (SRC) kinase is closely associated with decidualization and that SRC is indispensable for maximal decidualization in mice. To address whether SRC kinase activity is essential for decidualization in humans, hESCs were infected with adenoviruses carrying enhanced green fluorescent protein alone (Ad-EGFP), a kinase-inactive dominant-negative mutant (Ad-SRC/K295R), or an inactive autophosphorylation site mutant (Ad-SRC/Y416F). The cells were cultured in the presence of estradiol and progesterone (EP) to induce decidualization and subjected to RT-PCR, immunoblot, and ELISA analyses. Ad-EGFP-infected hESCs exhibited decidual transformation and up-regulation of decidualization markers including IGF binding protein 1 and prolactin in response to 12-d treatment with EP. In contrast, hESCs infected with Ad-SRC/K295R remained morphologically fibroblastoid without production of IGF binding protein 1 and prolactin even after EP treatment. Ad-SRC/Y416F displayed similar but less inhibitory effects on decidualization, compared with Ad-SRC/K295R. During decidualization, STAT5 was phosphorylated on tyrosine 694, a well-known SRC phosphorylation site. Phosphorylation was markedly attenuated by Ad-SRC/K295R but not Ad-EGFP. These results indicate that the SRC-STAT5 pathway is essential for decidualization of hESCs.


Assuntos
Diferenciação Celular/fisiologia , Decídua/patologia , Endométrio/patologia , Fator de Transcrição STAT5/metabolismo , Células Estromais/patologia , Quinases da Família src/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Decídua/efeitos dos fármacos , Decídua/metabolismo , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Fator de Crescimento Epidérmico/farmacologia , Estrogênios/farmacologia , Feminino , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Células NIH 3T3 , Fosforilação/efeitos dos fármacos , Progesterona/farmacologia , Prolactina/metabolismo , Transdução de Sinais/fisiologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo
14.
Proc Natl Acad Sci U S A ; 104(47): 18700-5, 2007 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18003928

RESUMO

Over the course of pregnancy, the human uterus undergoes a 500- to 1,000-fold increase in volume and a 24-fold increase in weight. The uterine smooth muscle layer or myometrium is remodeled, and both cell hypertrophy and hyperplasia are evident. The origin of the new smooth muscle cells, however, is unclear. They may arise from existing smooth muscle cells, or they may be the product of stem cell differentiation. This study describes a subset of myometrial cells isolated from nonpregnant human myometrium that represents the myometrial stem cell population. This was characterized as side population of myometrial cells (myoSP) by a distinct Hoechst dye efflux pattern. In contrast to the main population of myometrial cells (myoMP), myoSP resided in quiescence, underexpressed or lacked myometrial cell markers, and could proliferate and eventually differentiate into mature myometrial cells in vitro only under low oxygen concentration. Although myoMP displayed mature myometrial phenotypes before and after in vitro cultivation, only myoSP, not myoMP, generated functional human myometrial tissues efficiently when transplanted into the uteri of severely immunodeficient mice. Finally, myoSP were multipotent and made to differentiate into osteocytes and adipocytes in vitro under the appropriate differentiation-inducing conditions. Thus, myoSP exhibited phenotypic and functional characteristics of myometrial stem cells. Study of myoSP will improve the understanding of myometrial physiology and the pathogenesis of myometrium-derived diseases such as leiomyoma. myoSP may also represent a novel source of biological material that could be used in the reconstruction of not only the human uterus but also other organs as well.


Assuntos
Miométrio/citologia , Miométrio/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Adulto , Diferenciação Celular , Hipóxia Celular , Separação Celular , Feminino , Regulação da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Fenótipo
15.
Proc Natl Acad Sci U S A ; 104(6): 1925-30, 2007 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-17261813

RESUMO

Human uterine endometrium exhibits unique properties of cyclical regeneration and remodeling throughout reproductive life and also is subject to endometriosis through ectopic implantation of retrogradely shed endometrial fragments during menstruation. Here we show that functional endometrium can be regenerated from singly dispersed human endometrial cells transplanted beneath the kidney capsule of NOD/SCID/gamma(c)(null) immunodeficient mice. In addition to the endometrium-like structure, hormone-dependent changes, including proliferation, differentiation, and tissue breakdown and shedding (menstruation), can be reproduced in the reconstructed endometrium, the blood to which is supplied predominantly by human vessels invading into the mouse kidney parenchyma. Furthermore, the hormone-dependent behavior of the endometrium regenerated from lentivirally engineered endometrial cells expressing a variant luciferase can be assessed noninvasively and quantitatively by in vivo bioluminescence imaging. These results indicate that singly dispersed endometrial cells have potential applications for tissue reconstitution, angiogenesis, and human-mouse chimeric vessel formation, providing implications for mechanisms underlying the physiological endometrial regeneration during the menstrual cycle and the establishment of endometriotic lesions. This animal system can be applied as the unique model of endometriosis or for other various types of neoplastic diseases with the capacity of noninvasive and real-time evaluation of the effect of therapeutic agents and gene targeting when the relevant cells are transplanted beneath the kidney capsule.


Assuntos
Endométrio/fisiologia , Endométrio/transplante , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/cirurgia , Adulto , Animais , Endométrio/patologia , Feminino , Humanos , Rim/irrigação sanguínea , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Pessoa de Meia-Idade , Imunodeficiência Combinada Severa/patologia , Transplante Heterólogo/instrumentação , Transplante Heterólogo/métodos , Gravação em Vídeo
16.
Endocrinology ; 148(2): 896-902, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17068141

RESUMO

Histone deacetylase inhibitors (HDACIs) have recently emerged as promising anticancer drugs to induce cell cycle arrest, cytodifferentiation, and apoptosis. It is suggested, however, that HDACIs promote cell migration and invasion depending on the cell type. We have reported previously that treatment with HDACIs, including trichostatin A and suberoylanilide hydroxamic acid (SAHA) or progesterone in combination with estrogen, can induce cytodifferentiation of endometrial adenocarcinoma Ishikawa cells through up-regulation of glycodelin, a progesterone-induced endometrial glycoprotein. Given the reported role of glycodelin in cell motility and the migration-modulating potential of HDACIs, we investigated using wound healing assay and transwell migration assay whether ovarian steroid hormones, trichostatin A, or SAHA affects cell migration in endometrial cancer cell lines, Ishikawa and RL95-2. Treatment with ovarian steroid hormones, trichostatin A, and SAHA enhanced cell migration together with up-regulation of glycodelin. SAHA-augmented cell migration was almost completely blocked by gene silencing of glycodelin. Furthermore, overexpression of gycodelin alone resulted in increased cell motility in Ishikawa cells. Our results collectively indicate that glycodelin positively regulates cell motility acting as a mediator of HDACI-enhanced endometrial cell migration, suggesting the involvement of glycodelin in the dynamic endometrial gland morphogenesis during menstrual cycle. Our results raise a possibility that the use of HDACIs in the therapy for glycodelin-inducible endometrial and presumably other gynecological cancers may enhance invasion in cases in which the HDACIs fail to exert differentiation-inducing and/or antiproliferative effects.


Assuntos
Adenocarcinoma/fisiopatologia , Movimento Celular/efeitos dos fármacos , Neoplasias do Endométrio/fisiopatologia , Inibidores Enzimáticos/farmacologia , Glicoproteínas/metabolismo , Inibidores de Histona Desacetilases , Proteínas da Gravidez/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Feminino , Fibronectinas/metabolismo , Glicodelina , Humanos , Ácidos Hidroxâmicos/farmacologia , Ovário/metabolismo , Regulação para Cima , Vorinostat
17.
Hum Cell ; 19(1): 38-42, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16643606

RESUMO

Abstract Human uterine endometrium repeats proliferation, differentiation (decidualization) and tissue breakdown during the menstrual period. Appropriate secretion of ovarian steroid hormones regulates these sequential endometrial remodeling cycles. While progesterone replacement therapy is adopted for endometrial dysfunction of differentiation, including recurrent impairment of implantation, no obvious effective results are obtained. Histone reversible acetylation, regulated by histone acetyltransferases and histone deacetylases plays a pivotal role in gene transcription. Although, in cells cultured with histone deacetylase inhibitors (HDACI), the expression of only about 2% of expressed genes is changed twofold or more compared with untreated control cells. Numerous previous works have demonstrated that HDACI affect cell proliferation/apoptosis in a variety of types of cells. To date, several HDACI are in phase I or phase II clinical trials as anticancer drugs. However, no reports have been found that HDACI is useful for transdifferentiation in human endometrium. Recently, we reported that HDACI could induce the expression of differentiation marker proteins, morphological change and functional cytodifferentiation in both human endometrial stromal and epithelial cells. In this review, we summarize the effect of HDACI against the human endometrial cytodifferentiation, indicating the possibility that HDACI can be used not only as an anticancer drug, but also as a transdifferentiation reagent, based on a new strategy.


Assuntos
Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Endométrio/citologia , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/farmacologia , Acetilação , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Inibidores Enzimáticos/uso terapêutico , Feminino , Inativação Gênica/fisiologia , Glicodelina , Glicoproteínas/genética , Glicoproteínas/metabolismo , Histona Desacetilases/fisiologia , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/uso terapêutico , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Transcrição Gênica , Doenças Uterinas/tratamento farmacológico , Vorinostat
18.
Endocrinology ; 146(12): 5365-73, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16123169

RESUMO

Histone reversible acetylation, which is controlled by histone acetyltransferases and deacetylases, plays a fundamental role in gene transcription. Histone deacetylase inhibitors (HDACIs), such as trichostatin A (TSA) and suberoylanilide hydroxamic acid (SAHA), have been characterized not only as anticancer drugs, but also as cytodifferentiation-inducing agents. In human endometrium, postovulatory production of progesterone directs estrogen-primed endometrial glandular cells to differentiate and thereby produce a number of unique bioactive substances, including glycodelin, that are critical for implantation at the secretory phase of the menstrual cycle. In this study, we show that TSA and SAHA, belonging to the hydroxamic acid group of HDACIs, can induce the phenotype of a human endometrial adenocarcinoma cell line, Ishikawa (originally derived from the glandular component of the endometrium), to differentiate to closely resemble normal endometrial epithelium in a time- and dose-dependent manner, as determined by morphological changes, synthesis of glycogen, and expression of secretory phase-specific proteins, including glycodelin. The proliferation- and differentiation-modulating effects elicited by TSA and SAHA at their optimal concentrations were comparable or more potent than those exerted by combined treatment with progesterone and estradiol. Furthermore, the gene silencing of glycodelin by small interference RNA resulted in the blockade of HDACI-induced differentiation in Ishikawa cells, suggesting the requirement for glycodelin for endometrial epithelial differentiation. Our results collectively indicate that TSA and SAHA are potent differentiation inducers for endometrial glandular cells, providing a clue for a possible therapeutic strategy to modulate endometrial function by targeting glycodelin.


Assuntos
Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Inibidores Enzimáticos/farmacologia , Glicoproteínas/metabolismo , Inibidores de Histona Desacetilases , Proteínas da Gravidez/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Estradiol/administração & dosagem , Estradiol/farmacologia , Feminino , Inativação Gênica , Glicodelina , Glicogênio/biossíntese , Glicoproteínas/genética , Humanos , Ácidos Hidroxâmicos/farmacologia , Interleucina-6/biossíntese , Interleucina-6/genética , Fator Inibidor de Leucemia , Concentração Osmolar , Proteínas da Gravidez/genética , Progesterona/administração & dosagem , Progesterona/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/farmacologia , Regulação para Cima , Vorinostat
19.
Obstet Gynecol ; 105(5 Pt 2): 1215-8, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15863587

RESUMO

BACKGROUND: Enlarged multicystic ovaries and an elevated estradiol (E2) concentration have been reported as characteristics of follicle stimulating hormone (FSH)-secreting adenomas in reproductive-aged women. The natural course of the hormone in relationship to the microadenoma and ovarian findings, however, remains largely unknown. CASE: A 40-year-old woman with enlarged multicystic ovaries was nonsurgically diagnosed with an FSH-producing pituitary microadenoma. During her subsequent 12-month follow-up, the serum concentration of E2, but not FSH, and the size of the multicystic ovaries fluctuated dramatically. Both the E2 level and ovarian size were transiently normalized. CONCLUSION: Because of disease-related fluctuations, a reproductive-aged woman with an FSH-producing adenoma did not always present with laboratory values characteristic of ovarian hyperstimulation. This finding points out a possible pitfall in diagnosis and clinical management.


Assuntos
Adenoma/diagnóstico , Hormônio Foliculoestimulante/metabolismo , Neoplasias Hipofisárias/diagnóstico , Síndrome do Ovário Policístico/diagnóstico , Adenoma/complicações , Adenoma/terapia , Adulto , Endossonografia/métodos , Feminino , Seguimentos , Humanos , Imageamento por Ressonância Magnética/métodos , Neoplasias Hipofisárias/complicações , Neoplasias Hipofisárias/terapia , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/cirurgia , Medição de Risco , Índice de Gravidade de Doença
20.
Reprod Med Biol ; 4(2): 115-122, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29662388

RESUMO

Acetylation of histones is cooperatively regulated by two groups of enzymes, histone acetyltransferases and histone deacetylases. Histone acetylation status plays a fundamental role in the level of gene transcription; numerous studies have demonstrated that histone deacetylase inhibitors cause cell growth arrest, apoptosis, and differentiation in various cells including human mammary gland and endometrial cells by altering transcription of a small number of genes. A recent study has also shown that a highly acetylated histone status alters cell motility. After the present review of the published reports on the mechanisms underlying histone acetylation and in vitro effects of histone deacetylase inhibitors, we conclude that this class of agents may have potential not only as anticancer drugs, but also as inducers of differentiation and/or motility for benign gynecologic conditions such as endometriosis and disorders of endometrial differentiation and dysfunction. (Reprod Med Biol 2005; 4: 115-122).

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA