Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Biol Reprod ; 88(3): 80, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23349235

RESUMO

We have previously shown that the presence of trophoblast cells enhances invasiveness of decidualizing human endometrial stromal cells. The metastasis suppressor CD82, which has antimigratory function in tumor cells, is up-regulated in decidualizing endometrial stromal cells. CEACAM1 is expressed in trophoblast cells at the invasion front in early placenta and is considered proinvasive. Here, we investigate the role of CD82 and CEACAM1 in cocultures of the endometrial stromal cell line T-HESC and AC-1M88 trophoblast cells. In transwell migration assays, chemotaxis of AC-1M88 cells was stimulated by coplated T-HESC in the lower compartment or by the combination of heparin-binding EGF-like growth factor (HB-EGF), interleukin-1 beta (IL-1beta), and leukemia inhibitory factor (LIF), local factors present at the time of implantation. In an implantation model of AC-1M88 trophoblast spheroids on a monolayer of T-HESC, spheroid expansion was enhanced in the presence of HB-EGF/IL-1beta/LIF. Silencing of CEACAM1 in AC-1M88 blunted this response. Chemotactic migration of T-HESC was stimulated by trophoblast secretions or HB-EGF/IL-1beta/LIF. These responses were suppressed by CD82 depletion in T-HESC. Proteome profiling revealed the presence of platelet-derived growth factor (PDGF)-AA in trophoblast supernatant. Chemotaxis of T-HESC toward PDGF-AA was significantly inhibited by CD82 silencing. Neutralization of PDGF-AA in AC-1M88 conditioned media reduced the chemotactic effect on T-HESC. In summary, we demonstrate a mutual stimulation of chemotactic migration between trophoblast and endometrial stromal cells and promigratory roles for the cell surface molecules CEACAM1 and CD82, which may serve to support tissue remodeling at the implantation site.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Quimiotaxia , Endométrio/metabolismo , Proteína Kangai-1/metabolismo , Trofoblastos/metabolismo , Comunicação Celular , Linhagem Celular , Movimento Celular , Técnicas de Cocultura , Endométrio/citologia , Receptores ErbB/metabolismo , Feminino , Humanos , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteoma/metabolismo , Esferoides Celulares/fisiologia , Células Estromais/metabolismo
2.
Mol Hum Reprod ; 17(7): 421-33, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21357210

RESUMO

Successful pregnancy in humans depends on deep invasion of the maternal decidua by extravillous trophoblast cells (EVTs), a process regulated by autocrine and paracrine signals in the decidual-trophoblast microenvironment. Here we examined whether trophoblast invasion is affected by decidual differentiation of endometrial stromal cells (ESC) and modulated locally by cytokines and growth factors. Trophoblast spheroids were generated from the EVT-derived cell line AC-1M88 and placed onto monolayers of either undifferentiated or decidualized ESC, or directly onto tissue culture surface. Co-cultures were treated with epidermal growth factor (EGF), hepatocyte growth factor, heparin-binding EGF-like growth factor (HB-EGF), interleukin-1ß (IL-1ß) and leukaemia inhibitory factor (LIF). Expansion of spheroids over 2-3 days was significantly enhanced by a monolayer of undifferentiated ESC compared with tissue culture surface and further increased if ESC had been decidualized. HB-EGF and IL-1ß, alone or in combination with LIF, stimulated spheroid expansion but only on undifferentiated ESC. CEACAM1, an adhesion molecule implicated in trophoblast invasion, was up-regulated in AC-1M88 cells by conditioned medium from decidualized ESC, and by HB-EGF, IL-1ß and LIF in combination. Treatment of ESC with HB-EGF or IL-1ß increased the level of the tetraspanin CD82, a metastasis suppressor found in decidual cells at the implantation site. We suggest that decidualized ESC support trophoblast invasion by paracrine signals that may include HB-EGF, IL-1ß and LIF.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Interleucina-1beta/farmacologia , Trofoblastos/citologia , Western Blotting , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Endométrio/citologia , Fator de Crescimento Epidérmico/farmacologia , Feminino , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Imuno-Histoquímica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trofoblastos/efeitos dos fármacos
3.
Proc Natl Acad Sci U S A ; 105(39): 14928-33, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18809929

RESUMO

Evolutionary change in gene regulation can result from changes in cis-regulatory elements, leading to differences in the temporal and spatial expression of genes or in the coding region of transcription factors leading to novel functions or both. Although there is a growing body of evidence supporting the importance of cis-regulatory evolution, examples of protein-mediated evolution of novel developmental pathways have not been demonstrated. Here, we investigate the evolution of prolactin (PRL) expression in endometrial cells, which is essential for placentation/pregnancy in eutherian mammals and is a direct regulatory target of the transcription factor HoxA-11. Here, we show that (i) endometrial PRL expression is a derived feature of placental mammals, (ii) the PRL regulatory gene HoxA-11 experienced a period of strong positive selection in the stem-lineage of eutherian mammals, and (iii) only HoxA-11 proteins from placental mammals, including the reconstructed ancestral eutherian gene, are able to up-regulate PRL from the promoter used in endometrial cells. In contrast, HoxA-11 from the reconstructed therian ancestor, opossum, platypus, and chicken are unable to up-regulate PRL expression. These results demonstrate that the evolution of novel gene expression domains is not only mediated by the evolution of cis-regulatory elements but can also require evolutionary changes of transcription factor proteins themselves.


Assuntos
Endométrio/metabolismo , Evolução Molecular , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Gravidez/genética , Prolactina/genética , Sequência de Aminoácidos , Animais , Galinhas/genética , Galinhas/metabolismo , Elefantes/genética , Elefantes/metabolismo , Feminino , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Gambás/genética , Gambás/metabolismo , Placenta/metabolismo , Regiões Promotoras Genéticas , Seleção Genética
4.
Reprod Biol Endocrinol ; 7: 76, 2009 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-19619280

RESUMO

BACKGROUND: Coordinated differentiation of the endometrial compartments in the second half of the menstrual cycle is a prerequisite for the establishment of pregnancy. Endometrial stromal cells (ESC) decidualize under the influence of ovarian progesterone to accommodate implantation of the blastocyst and support establishment of the placenta. Studies into the mechanisms of decidualization are often hampered by the lack of primary ESC. Here we describe a novel immortalized human ESC line. METHODS: Primary ESC were immortalized by the transduction of telomerase. The resultant cell line, termed St-T1b, was characterized for its morphological and biochemical properties by immunocytochemistry, RT-PCR and immunoblotting. Its progestational response was tested using progesterone and medroxyprogesterone acetate with and without 8-Br-cAMP, an established inducer of decidualization in vitro. RESULTS: St-T1b were positive for the fibroblast markers vimentin and CD90 and negative for the epithelial marker cytokeratin-7. They acquired a decidual phenotype indistinguishable from primary ESC in response to cAMP stimulation. The decidual response was characterized by transcriptional activation of marker genes, such as PRL, IGFBP1, and FOXO1, and enhanced protein levels of the tumor suppressor p53 and the metastasis suppressor KAI1 (CD82). Progestins alone had no effect on St-T1b cells, but medroxyprogesterone acetate greatly enhanced the cAMP-stimulated expression of IGFBP-1 after 3 and 7 days. Progesterone, albeit more weakly, also augmented the cAMP-induced IGFBP-1 production but only after 7 days of treatment. The cell line remained stable in continuous culture for more than 150 passages. CONCLUSION: St-T1b express the appropriate phenotypic ESC markers and their decidual response closely mimics that of primary cultures. Decidualization is efficiently induced by cAMP analog and enhanced by medroxyprogesterone acetate, and, to a lesser extent, by natural progesterone. St-T1b cells therefore serve as a useful model for primary ESC.


Assuntos
Linhagem Celular , Endométrio/citologia , Células Estromais/citologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Adulto , Decídua/fisiologia , Receptor alfa de Estrogênio/biossíntese , Feminino , Humanos , Acetato de Medroxiprogesterona/farmacologia , Pessoa de Meia-Idade , Progesterona/farmacologia , Receptores de Progesterona/biossíntese , Células Estromais/efeitos dos fármacos , Telomerase/genética , Transdução Genética
5.
Endocrinology ; 149(11): 5872-87, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18617611

RESUMO

Functional progesterone withdrawal associated with human parturition has been ascribed to various mechanisms modulating the function of the classical progesterone receptors (PRs), B and A, in utero. These include up-regulation of the inhibitory PR-C isoform, described as a 60-kDa protein occurring from translation initiation at codon 595. Our initial attempts to detect PR-C yielded uninterpretable results. To systematically validate antibodies for immunodetection of PR isoforms, we generated expression vectors for PR variants originating from putative start codons AUG-289, -301, -595, -632, and -692 in addition to those for PR-B and PR-A, and for alternative splice variants PR-T, PR-S, and PR-M. All constructs were subjected to in vitro and in vivo translation and immunoblotting with a panel of 13 PR antibodies. Antibodies raised against full-length PR were generally not capable of detecting N-terminally truncated forms, whereas C-terminal antibodies did not or only weakly reacted with PR-B and PR-A but produced prominent nonspecific signals. Thus, immunodetection of N-terminally truncated PR isoforms is prone to artifacts. Proteins of about 64 kDa were expressed from PR-289 and -301, but no corresponding endogenous forms were observed. PR-T, PR-S, and PR-M cDNAs yielded no detectable translation products. No protein was translated from AUG-595 in our PR-C expression vector unless a Kozak sequence was introduced, and the product was not 60 but 38 kDa in size. Thus, the 60-kDa protein called PR-C does not originate from AUG-595 and is not a naturally occurring PR isoform.


Assuntos
Anticorpos/metabolismo , Perfilação da Expressão Gênica , Receptores de Progesterona/genética , Receptores de Progesterona/imunologia , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Feminino , Humanos , Imunoensaio , Miométrio/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/isolamento & purificação , Isoformas de Proteínas/metabolismo , Sítios de Splice de RNA/genética , Sítios de Splice de RNA/fisiologia , Receptores de Progesterona/isolamento & purificação , Receptores de Progesterona/metabolismo , Sítio de Iniciação de Transcrição/fisiologia
6.
Steroids ; 73(9-10): 942-52, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18221973

RESUMO

The recent discovery of three closely related cell surface receptors that bind to progesterone and mediate its actions on various cytoplasmic signalling cascades has been heralded as a major break-through. The reason for this is all too obvious. Progesterone is an essential regulator of all major reproductive events and progestins and antiprogestins are widely used in the treatment of many different gynaecological and obstetrical disorders. The novel membrane progestin receptors (mPRalpha, beta, gamma) reportedly resemble and function as G-protein-coupled receptors and therefore are promising pharmaceutical targets. However, our studies failed to corroborate that mPRs are expressed on the cell surface, that they mediate rapid progesterone signalling events, and even that they are bona fide progestin binding moieties. While the reason for these startling opposing results remains unclear, a critical review of existing data may help to shed some light onto the controversial mPRs. Time has come to talk.


Assuntos
Regulação da Expressão Gênica , Receptores de Progesterona/metabolismo , Animais , Membrana Celular/metabolismo , Estruturas Celulares , Citoplasma/metabolismo , Feminino , Humanos , Ligantes , Modelos Biológicos , Progesterona/metabolismo , Regiões Promotoras Genéticas , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
7.
Semin Reprod Med ; 25(6): 445-53, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17960529

RESUMO

The fetomaternal interface, consisting of the maternal decidua and the invading fetal trophoblast, critically regulates placental function and the growth and development of the conceptus. In its broadest sense, decidualization could be viewed as the postovulatory process of endometrial remodeling in preparation for pregnancy, which includes secretory transformation of the uterine glands, influx of specialized uterine natural killer cells, and vascular remodeling. A more restricted definition of the decidual process denotes the morphological and biochemical reprogramming of the endometrial stromal compartment. This differentiation process is dependent entirely on the convergence of the cyclic adenosine monophosphate and progesterone signaling pathways that drives integrated changes at both the transcriptome and the proteome level. As a consequence, decidualizing stromal cells acquire the unique ability to regulate trophoblast invasion, to resist inflammatory and oxidative insults, and to dampen local maternal immune responses. In humans, decidualization of the stromal compartment occurs in the mid-luteal phase of the menstrual cycle, independently of pregnancy. This raises the possibility that biochemical analysis of timed endometrial biopsy samples taken in a nonconception cycle could be informative of subsequent pregnancy outcome.


Assuntos
Decídua/metabolismo , Implantação do Embrião , Embrião de Mamíferos/metabolismo , Endométrio/metabolismo , Placentação , Transdução de Sinais , Adesão Celular , Gonadotropina Coriônica/metabolismo , AMP Cíclico/metabolismo , Decídua/citologia , Decídua/imunologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/imunologia , Endométrio/citologia , Endométrio/imunologia , Feminino , Humanos , Infertilidade/metabolismo , Infertilidade/fisiopatologia , Estresse Oxidativo , Gravidez , Complicações na Gravidez/metabolismo , Complicações na Gravidez/fisiopatologia , Progesterona/metabolismo , Células Estromais/metabolismo , Trofoblastos/metabolismo
8.
J Androl ; 28(1): 59-66, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-16899810

RESUMO

Due to alternative promoter usage, splicing, and translational initiation, expression of the cAMP-responsive element modulator (CREM) gene results in the production of functionally different CREM proteins with either activating or repressing potential on target gene expression. Recently, we demonstrated 2 novel isoforms (CREM-2-F-G-H-Ib and CREM-2-G-H-Ib) in various germ cell types during normal and impaired human spermatogenesis. In contrast to known isoforms, these exhibit a transactivation domain but lack a kinase-inducible domain (KID) domain resulting in a disruption of the open reading frame. In the present study, we functionally analyzed these isoforms. Investigation of both in vitro and in vivo expressed proteins from human testis RNA suggests that a novel upstream open reading frame in exon 2 is translated from isoform CREM-2-F-G-H-Ib, giving rise to a full-length protein. Furthermore, in both isoforms, usage of downstream adeninethymine-guanines (ATGs) for translation initiation could be observed. Sequence-specific DNA binding of CREM isoforms was confirmed by electrophoretic mobility shift assays. Luciferase reporter gene assays in cells transfected with novel CREM cDNAs demonstrated that protein kinase A dependent stimulation was inhibited by coexpression of CREM-2-F-G-H-Ib but not of CREM-2-G-H-Ib.


Assuntos
Modulador de Elemento de Resposta do AMP Cíclico/fisiologia , Espermatozoides/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Western Blotting , Linhagem Celular Tumoral , Clonagem Molecular , Modulador de Elemento de Resposta do AMP Cíclico/química , Modulador de Elemento de Resposta do AMP Cíclico/genética , DNA/metabolismo , Humanos , Masculino , Dados de Sequência Molecular , Fases de Leitura Aberta , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Transcrição Gênica
9.
Mol Endocrinol ; 20(12): 3146-64, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16959873

RESUMO

The steroid hormone progesterone exerts pleiotrophic functions in many cell types. Although progesterone controls transcriptional activation through binding to its nuclear receptors, it also initiates rapid nongenomic signaling events. Recently, three putative membrane progestin receptors (mPRalpha, beta, and gamma) with structural similarity to G protein-coupled receptors have been identified. These mPR isoforms are expressed in a tissue-specific manner and belong to the larger, highly conserved family of progestin and adiponectin receptors found in plants, eubacteria, and eukaryotes. The fish mPRalpha has been reported to mediate progesterone-dependent MAPK activation and inhibition of cAMP production through coupling to an inhibitory G protein. To functionally characterize the human homologs, we established human embryonic kidney 293 and MDA-MB-231 cell lines that stably express human mPRalpha, beta, or gamma. For comparison, we also established cell lines expressing the mPRalpha cloned from the spotted seatrout (Cynoscion nebulosus) and Japanese pufferfish (Takifugu rubripes). Surprisingly, we found no evidence that human or fish mPRs regulate cAMP production or MAPK (ERK1/2 or p38) activation upon progesterone stimulation. Furthermore, the mPRs did not couple to a highly promiscuous G protein subunit, Galpha(q5i), in transfection studies or provoke Ca(2+) mobilization in response to progesterone. Finally, we demonstrate that transfected mPRs, as well as endogenous human mPRalpha, localize to the endoplasmic reticulum, and that their expression does not lead to increased progestin binding either in membrane preparations or in intact cells. Our results therefore do not support the concept that mPRs are plasma membrane receptors involved in transducing nongenomic progesterone actions.


Assuntos
Retículo Endoplasmático/química , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de Progesterona/análise , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Linhagem Celular , Retículo Endoplasmático/metabolismo , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Perciformes/metabolismo , Filogenia , Progesterona/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Progesterona/agonistas , Receptores de Progesterona/metabolismo , Takifugu/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Mol Endocrinol ; 16(1): 141-54, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11773445

RESUMO

Activation of the decidual PRL (dPRL) promoter, a major differentiation marker in human endometrial stromal (ES) cells, by cAMP is effected through the induction and binding of CCAAT/enhancer-binding protein-beta (C/EBPbeta) to two overlapping cognate response elements in the promoter region dPRL-332/-270. Progesterone is essential for decidualization and potently enhances cAMP-dependent dPRL promoter activity. We now demonstrate that both liganded PR isoforms, PR-A and PR-B, are capable of trans-activating the dPRL-332/-270 region. The absence of a palindromic progesterone response element (PRE) within this promoter region suggested cross-coupling between C/EBPbeta and PR in human ES cells. Physical interaction between these distinct transcription factors was confirmed by glutathione-S-transferase pull-down assays, demonstrating that both C/EBPbeta isoforms, the full-length activator liver-enriched activatory protein (LAP) and the truncated inhibitor liver-enriched inhibitory protein (LIP), can bind PR-B as well as PR-A in vitro. Transient transfection studies in primary ES cells were used to examine the consequences of PR and C/EBPbeta interaction on activation of their respective response elements. Activation of mouse mammary tumor virus promoter or a reporter construct containing two isolated palindromic PREs by liganded PR-B was synergistically enhanced by coexpression of LIP, but not LAP. In contrast, PR-A failed to trans-activate these constructs significantly regardless of the presence of either C/EBPbeta isoform. Conversely, LAP-dependent activation of the dPRL-332/-270 region or a reporter construct driven by a single C/EBPbeta response element was greatly enhanced by PR-A, but not PR-B, in a ligand-dependent manner. These observations reveal that PR and C/EBPbeta isoform ratios are important determinants of the cellular response to ovarian progesterone in the reproductive tract; the predominance of PR-A and LAP favors expression of C/EBPbeta-dependent genes, whereas PR-B and LIP cooperate in activating PRE-driven promoters.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Endométrio/citologia , Endométrio/metabolismo , Prolactina/metabolismo , Sequência de Bases , Sítios de Ligação , Proteína beta Intensificadora de Ligação a CCAAT/genética , Células Cultivadas , Feminino , Humanos , Dados de Sequência Molecular , Mutação , Prolactina/genética , Regiões Promotoras Genéticas , Isoformas de Proteínas , Elementos de Resposta , Células Estromais/metabolismo , Transcrição Gênica , Ativação Transcricional
11.
J Clin Endocrinol Metab ; 89(10): 5233-44, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15472230

RESUMO

Decidualization of the endometrial stromal compartment is critical for embryo implantation. Initiation of this differentiation process requires elevated intracellular cAMP levels. We now report a massive and sustained up-regulation of p53 tumor suppressor protein during cAMP-induced decidualization of cultured endometrial stromal cells. Nuclear accumulation of p53 was not accompanied by increased mRNA expression, suggesting stabilization of the protein as the underlying mechanism. Proteasomal degradation of p53 is known to be mediated by nuclear Mdm2. Nuclear translocation of Mdm2, in turn, is dependent on phosphorylation by protein kinase B/Akt (PKB/Akt). In cAMP-treated decidualized cells, p53 accumulation was associated with decreased nuclear Mdm2 and cytoplasmic PKB/Akt levels. Conversely, withdrawal of the decidualization stimulus resulted in morphological and biochemical dedifferentiation, disappearance of p53, but increased abundance of PKB/Akt. Furthermore, Western blot and immunohistochemical analyses of endometrial biopsies confirmed that p53 is expressed in vivo in the stromal compartment during the late secretory phase of the cycle. The observation that p53 protein expression is closely associated with decidual transformation indicates a novel role for this tumor suppressor in regulating human endometrial function.


Assuntos
AMP Cíclico/metabolismo , Decídua/citologia , Decídua/fisiologia , Células Estromais/fisiologia , Proteína Supressora de Tumor p53/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Humanos , Regiões Promotoras Genéticas/fisiologia , RNA Mensageiro/metabolismo , Células Estromais/citologia , Proteína Supressora de Tumor p53/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Regulação para Cima , Leveduras
12.
Virchows Arch ; 444(2): 149-52, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14691721

RESUMO

The human trophoblast has the capacity to invade maternal tissue in a controlled fashion and to produce a wide range of hormones. The transcription factors belonging to the CCAAT/enhancer-binding protein (C/EBP) family are regulators of intracellular processes and mediators of hormone action. C/EBP binding sites have been described in the promoters of several placenta-expressed target genes. In the present study, we used immunohistochemistry and Western-blot analysis to investigate the expression pattern of the three most important members of this family, C/EBP-alpha, -beta, and -delta, in the normal human placenta as well as in isolated trophoblast cell populations. We found C/EBP-alpha and C/EBP-beta expression in the villous syncytiotrophophoblast (ST) and the extravillous (intermediate) trophoblast (EVT), but it was absent from the villous cytotrophoblast (CT). Interestingly, expression of C/EBP-beta continued to be very strong up to the third trimester of pregnancy, especially in the ST. C/EBP-delta showed overall lower expression levels, stronger only in the EVT, while CT/ST showed very low/negative expression. These data show for the first time the expression pattern and tissue localization of C/EBP factors in the human placenta, indicating that these factors (especially C/EBP-beta) may play important roles in the regulation of placenta-specific genes and processes.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/biossíntese , Placenta/metabolismo , Western Blotting , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Gravidez , Trimestres da Gravidez , Trofoblastos/metabolismo
13.
Endocr Rev ; 35(6): 851-905, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25141152

RESUMO

Decidualization denotes the transformation of endometrial stromal fibroblasts into specialized secretory decidual cells that provide a nutritive and immunoprivileged matrix essential for embryo implantation and placental development. In contrast to most mammals, decidualization of the human endometrium does not require embryo implantation. Instead, this process is driven by the postovulatory rise in progesterone levels and increasing local cAMP production. In response to falling progesterone levels, spontaneous decidualization causes menstrual shedding and cyclic regeneration of the endometrium. A growing body of evidence indicates that the shift from embryonic to maternal control of the decidual process represents a pivotal evolutionary adaptation to the challenge posed by invasive and chromosomally diverse human embryos. This concept is predicated on the ability of decidualizing stromal cells to respond to individual embryos in a manner that either promotes implantation and further development or facilitates early rejection. Furthermore, menstruation and cyclic regeneration involves stem cell recruitment and renders the endometrium intrinsically capable of adapting its decidual response to maximize reproductive success. Here we review the endocrine, paracrine, and autocrine cues that tightly govern this differentiation process. In response to activation of various signaling pathways and genome-wide chromatin remodeling, evolutionarily conserved transcriptional factors gain access to the decidua-specific regulatory circuitry. Once initiated, the decidual process is poised to transit through distinct phenotypic phases that underpin endometrial receptivity, embryo selection, and, ultimately, resolution of pregnancy. We discuss how disorders that subvert the programming, initiation, or progression of decidualization compromise reproductive health and predispose for pregnancy failure.


Assuntos
AMP Cíclico/fisiologia , Decídua/fisiologia , Endométrio/fisiologia , Ciclo Menstrual/fisiologia , Progesterona/fisiologia , Diferenciação Celular/fisiologia , Decídua/citologia , Implantação do Embrião/fisiologia , Endométrio/citologia , Feminino , Humanos , Gravidez , Saúde Reprodutiva
14.
Hum Reprod Update ; 19(5): 542-57, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23827985

RESUMO

BACKGROUND Mechanisms underlying early reproductive loss in the human are beginning to be elucidated. The migratory and invasive capacity of human endometrial stromal cells (ESCs) is increasingly recognized to contribute to the intense tissue remodelling associated with embryo implantation, trophoblast invasion and endometrial regeneration. In this review, we examine the signals and mechanisms that control ESC migration and invasion and assess how deregulation of these cell functions contributes to common reproductive disorders. METHODS The PubMed database was searched for publications on motility and invasiveness of human ESCs in normal endometrial function and in reproductive disorders including implantation failure, recurrent pregnancy loss (RPL), endometriosis and adenomyosis, covering the period 2000-2012. RESULTS Increasing evidence suggests that implantation failure and RPL involve abnormal migratory responses of decidualizing ESCs to embryo and trophoblast signals. Numerous reports indicate that endometriosis, as well as adenomyosis, is associated with increased basal and stimulated invasiveness of ESCs and their progenitor cells, suggesting a link between a heightened menstrual repair response and the formation of ectopic implants. Migration and invasiveness of ESCs are controlled by a complex array of hormones, growth factors, chemokines and inflammatory mediators, and involve signalling through Rho GTPases, phosphatidylinositol-3-kinase and mitogen-activated protein kinase pathways. CONCLUSIONS Novel concepts are extending our understanding of the key functions of ESCs in effecting tissue repair imposed by cyclic menstruation and parturition. Migration of decidualizing ESCs also serves to support blastocyst implantation and embryo selection through discriminate motile responses directed by embryo quality. Targeting regulatory molecules holds promise for developing new strategies for the treatment of reproductive disorders such as endometriosis and recurrent miscarriage; and harnessing the migratory capacity of progenitor mesenchymal stem cells in the endometrium may offer new opportunities in regenerative medicine.


Assuntos
Movimento Celular , Endométrio/citologia , Aborto Habitual/patologia , Adenomiose/patologia , Implantação do Embrião/fisiologia , Endometriose/patologia , Endométrio/fisiologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Gravidez , Regeneração , Transdução de Sinais , Células-Tronco/patologia , Células Estromais/patologia , Células Estromais/fisiologia , Trofoblastos/metabolismo , Trofoblastos/fisiologia
15.
PLoS One ; 8(1): e54336, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23349855

RESUMO

Human implantation involves extensive tissue remodeling at the fetal-maternal interface. It is becoming increasingly evident that not only trophoblast, but also decidualizing endometrial stromal cells are inherently motile and invasive, and likely contribute to the highly dynamic processes at the implantation site. The present study was undertaken to further characterize the mechanisms involved in the regulation of endometrial stromal cell motility and to identify trophoblast-derived factors that modulate migration. Among local growth factors known to be present at the time of implantation, heparin-binding epidermal growth factor-like growth factor (HB-EGF) triggered chemotaxis (directed locomotion), whereas platelet-derived growth factor (PDGF)-BB elicited both chemotaxis and chemokinesis (non-directed locomotion) of endometrial stromal cells. Supernatants of the trophoblast cell line AC-1M88 and of first trimester villous explant cultures stimulated chemotaxis but not chemokinesis. Proteome profiling for cytokines and angiogenesis factors revealed neither PDGF-BB nor HB-EGF in conditioned media from trophoblast cells or villous explants, while placental growth factor, vascular endothelial growth factor and PDGF-AA were identified as prominent secretory products. Among these, only PDGF-AA triggered endometrial stromal cell chemotaxis. Neutralization of PDGF-AA in trophoblast conditioned media, however, did not diminish chemoattractant activity, suggesting the presence of additional trophoblast-derived chemotactic factors. Pathway inhibitor studies revealed ERK1/2, PI3 kinase/Akt and p38 signaling as relevant for chemotactic motility, whereas chemokinesis depended primarily on PI3 kinase/Akt activation. Both chemotaxis and chemokinesis were stimulated upon inhibition of Rho-associated, coiled-coil containing protein kinase. The chemotactic response to trophoblast secretions was not blunted by inhibition of isolated signaling cascades, indicating activation of overlapping pathways in trophoblast-endometrial communication. In conclusion, trophoblast signals attract endometrial stromal cells, while PDGF-BB and HB-EGF, although not identified as trophoblast-derived, are local growth factors that may serve to fine-tune directed and non-directed migration at the implantation site.


Assuntos
Movimento Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteínas Proto-Oncogênicas c-sis/farmacologia , Células Estromais/efeitos dos fármacos , Becaplermina , Western Blotting , Linhagem Celular , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Vilosidades Coriônicas/metabolismo , Endométrio/citologia , Receptores ErbB/metabolismo , Feminino , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Crescimento Placentário , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Gravidez , Proteínas da Gravidez/metabolismo , Proteínas da Gravidez/farmacologia , Primeiro Trimestre da Gravidez , Proteoma/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Estromais/metabolismo , Técnicas de Cultura de Tecidos , Trofoblastos/citologia , Trofoblastos/metabolismo , Quinases Associadas a rho/metabolismo
16.
PLoS One ; 7(7): e41424, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22848492

RESUMO

BACKGROUND: The aetiology of recurrent miscarriage (RM) remains largely unexplained. Women with RM have a shorter time to pregnancy interval than normally fertile women, which may be due to more frequent implantation of non-viable embryos. We hypothesized that human endometrial stromal cells (H-EnSCs) of women with RM discriminate less effectively between high-and low-quality human embryos and migrate more readily towards trophoblast spheroids than H-EnSCs of normally fertile women. METHODOLOGY/PRINCIPAL FINDINGS: Monolayers of decidualized H-EnSCs were generated from endometrial biopsies of 6 women with RM and 6 fertile controls. Cell-free migration zones were created and the effect of the presence of a high-quality (day 5 blastocyst, n = 13), a low-quality (day 5 blastocyst with three pronuclei or underdeveloped embryo, n = 12) or AC-1M88 trophoblast cell line spheroid on H-ESC migratory activity was analyzed after 18 hours. In the absence of a spheroid or embryo, migration of H-EnSCs from fertile or RM women was similar. In the presence of a low-quality embryo in the zone, the migration of H-EnSCs of control women was inhibited compared to the basal migration in the absence of an embryo (P<0.05) and compared to the migration in the presence of high-quality embryo (p<0.01). Interestingly, the migratory response H-EnSCs of women with RM did not differ between high- and low-quality embryos. Furthermore, in the presence of a spheroid their migration was enhanced compared to the H-EnSCs of controls (p<0.001). CONCLUSIONS: H-EnSCs of fertile women discriminate between high- and low-quality embryos whereas H-EnSCs of women with RM fail to do so. H-EnSCs of RM women have a higher migratory response to trophoblast spheroids. Future studies will focus on the mechanisms by which low-quality embryos inhibit the migration of H-EnSCs and how this is deregulated in women with RM.


Assuntos
Aborto Habitual/metabolismo , Movimento Celular , Endométrio/metabolismo , Trofoblastos/metabolismo , Aborto Habitual/patologia , Aborto Habitual/fisiopatologia , Adulto , Células Cultivadas , Endométrio/patologia , Feminino , Humanos , Gravidez , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Células Estromais/metabolismo , Células Estromais/patologia , Trofoblastos/patologia
17.
PLoS One ; 4(9): e6845, 2009 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-19727442

RESUMO

BACKGROUND: During the menstrual cycle, the ovarian steroid hormones estrogen and progesterone control a dramatic transcriptional reprogramming of endometrial stromal cells (ESCs) leading to a receptive state for blastocyst implantation and the establishment of pregnancy. A key marker gene of this decidualization process is the prolactin gene. Several transcriptional regulators have been identified that are essential for decidualization of ESCs, including the Hox genes HoxA-10 and HoxA-11, and the forkhead box gene FOXO1A. While previous studies have identified downstream target genes for HoxA-10 and FOXO1A, the role of HoxA-11 in decidualization has not been investigated. Here, we show that HoxA-11 is required for prolactin expression in decidualized ESC. While HoxA-11 alone is a repressor on the decidual prolactin promoter, it turns into an activator when combined with FOXO1A. Conversely, HoxA-10, which has been previously shown to associate with FOXO1A to upregulate decidual IGFBP-1 expression, is unable to upregulate PRL expression when co-expressed with FOXO1A. By co-immunoprecipitation and chromatin immunoprecipitation, we demonstrate physical association of HoxA-11 and FOXO1A, and binding of both factors to an enhancer region (-395 to -148 relative to the PRL transcriptional start site) of the decidual prolactin promoter. Because FOXO1A is induced upon decidualization, it serves to assemble a decidual-specific transcriptional complex including HoxA-11. These data highlight cooperativity between numerous transcription factors to upregulate PRL in differentiating ESC, and suggest that this core set of transcription factors physically and functionally interact to drive the expression of a gene battery upregulated in differentiated ESC. In addition, the functional non-equivalence of HoxA-11 and HoxA-10 with respect to PRL regulation suggests that these transcription factors regulate distinct sets of target genes during decidualization.


Assuntos
Decídua/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Prolactina/biossíntese , Animais , Sítios de Ligação , Endométrio/metabolismo , Feminino , Proteína Forkhead Box O1 , Genes Reporter , Células HeLa , Humanos , Progesterona/metabolismo , Prolactina/metabolismo , Regiões Promotoras Genéticas , Transcrição Gênica
19.
Am J Pathol ; 170(1): 126-39, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17200188

RESUMO

At the human maternal-fetal interface, the decidua forms a dense matrix that is believed to limit trophoblast invasion. We investigated whether the metastasis suppressor KAI1 (CD82) is expressed at the maternal-fetal interface. Immunohistochemistry showed strong expression of KAI1 in decidual cells, whereas trophoblast cells were negative for KAI1. In luteal phase endometrium, KAI1 was present in decidualizing endometrial stromal cells. We investigated whether KAI1 expression in endometrial stromal cells is regulated by the decidualizing stimuli cAMP and progesterone or by the cytokine interleukin (IL)-1beta. Western blot analysis revealed induction of KAI1 protein by cAMP analog, but not by progesterone, in a delayed fashion. In contrast, IL-1beta rapidly stimulated KAI1 expression at the transcript level and at the protein level. Cultured decidual cells from term placenta expressed a basal level of KAI1 protein that was elevated on cAMP stimulation. Silencing of KAI1 by RNA interference attenuated expression of decorin, a decidual product implicated in limiting trophoblast invasion. This study shows for the first time the expression of KAI1 in decidual cells at the human maternal-fetal interface, where the metastasis suppressor might participate in intercellular communication with trophoblast cells and the control of trophoblast invasion.


Assuntos
Decídua/metabolismo , Proteína Kangai-1/fisiologia , Comunicação Celular , Movimento Celular , Células Cultivadas , Decídua/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imuno-Histoquímica , Gravidez , Trofoblastos/metabolismo , Proteínas Supressoras de Tumor/fisiologia
20.
J Biol Chem ; 281(1): 269-78, 2006 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-16227626

RESUMO

The tumor suppressor protein p53 is not only involved in defending cells against genotoxic insults but is also implicated in differentiation processes, a function that it shares with the CCAAT/enhancer-binding protein beta (C/EBPbeta). We previously reported an up-regulation of both factors in the cycle-dependent differentiation process of human endometrial stromal cells, termed decidualization. C/EBPbeta-mediated activation of a decidualization marker, the decidual prolactin promoter, was antagonized by p53. Here we report that C/EBPbeta in turn represses the transcriptional activity of p53. Competition for limiting amounts of coactivator CREB-binding protein/p300 was ruled out as the underlying mechanism of transrepression. Physical interaction between p53 and C/EBPbeta was demonstrated in vitro and in vivo and shown to depend on the C-terminal domains of both proteins. In gel shift experiments, C/EBPbeta reduced complex formation between p53 and its response element. Conversely, p53 strongly inhibited binding of endogenous C/EBPbeta from endometrial stromal cells to the C/EBP-responsive region in the decidual prolactin promoter. The observed negative cross-talk between p53 and C/EBPbeta is likely to impact expression of their respective target genes.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Regulação da Expressão Gênica/fisiologia , Células Estromais/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Ensaio de Desvio de Mobilidade Eletroforética , Endométrio/citologia , Feminino , Humanos , Osteossarcoma , Elementos de Resposta/fisiologia , Células Estromais/citologia , Fatores de Transcrição de p300-CBP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA