Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 42(38): 2854-2867, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37587334

RESUMO

Increasing evidence points towards a causal link between exposure to persistent organic pollutants (POPs) with increased incidence and aggressivity of various cancers. Among these POPs, dioxin and PCB-153 are widely found in our environment and represent a significant source of contamination. Dioxin exposure has already been linked to cancer such as non-Hodgkin's lymphoma, but remains to be more extensively investigated in other cancers. Potential implications of dioxin and PCB-153 in prostate cancer progression spurred us to challenge both ex vivo and in vivo models with low doses of these POPs. We found that dioxin or PCB-153 exposure increased hallmarks of growth and metastasis of prostate cancer cells ex vivo and in grafted NOD-SCID mice. Exposure induced histopathological carcinoma-like patterns in the Ptenpc-/- mice. We identified up-regulation of Acetyl-CoA Acetyltransferase-1 (ACAT1) involved in ketone bodies pathway as a potential target. Mechanistically, genetic inhibition confirmed that ACAT1 mediated dioxin effect on cell migration. Using public prostate cancer datasets, we confirmed the deregulation of ACAT1 and associated gene encoded ketone bodies pathway enzymes such as OXCT1, BDH1 and HMGCL in advanced prostate cancer. To further explore this link between dioxin and ACAT1 deregulation, we analyzed a unique prostate-tumour tissue collection from the USA veterans exposed to agent orange, known to be highly contaminated by dioxin because of industrial production. We found that ACAT1 histoscore is significantly increased in exposed patients. Our studies reveal the implication of dioxin and PCB-153 to induce a prometastatic programme in prostate tumours and identify ACAT1 deregulation as a key event in this process.


Assuntos
Dioxinas , Dibenzodioxinas Policloradas , Neoplasias da Próstata , Masculino , Humanos , Animais , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Poluentes Orgânicos Persistentes , Dioxinas/toxicidade , Neoplasias da Próstata/induzido quimicamente , Neoplasias da Próstata/genética , Acetiltransferases
2.
Int J Mol Sci ; 23(23)2022 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-36499726

RESUMO

Understanding the regulation of the testicular endocrine function leading to testosterone production is a major objective as the alteration of endocrine function is associated with the development of many diseases such as infertility. In the last decades, it has been demonstrated that several endogenous molecules regulate the steroidogenic pathway. Among them, bile acids have recently emerged as local regulators of testicular physiology and particularly endocrine function. Bile acids act through the nuclear receptor FXRα (Farnesoid-X-receptor alpha; NR1H4) and the G-protein-coupled bile acid receptor (GPBAR-1; TGR5). While FXRα has been demonstrated to regulate testosterone synthesis within Leydig cells, no data are available regarding TGR5. Here, we investigated the potential role of TGR5 within Leydig cells using cell culture approaches combined with pharmacological exposure to the TGR5 agonist INT-777. The data show that activation of TGR5 results in a decrease in testosterone levels. TGR5 acts through the PKA pathway to regulate steroidogenesis. In addition, our data show that TGR5 activation leads to an increase in cholesterol ester levels. This suggests that altered lipid homeostasis may be a mechanism explaining the TGR5-induced decrease in testosterone levels. In conclusion, the present work highlights the impact of the TGR5 signaling pathway on testosterone production and reinforces the links between bile acid signaling pathways and the testicular endocrine function. The testicular bile acid pathways need to be further explored to increase our knowledge of pathologies associated with impaired testicular endocrine function, such as fertility disorders.


Assuntos
Ácidos e Sais Biliares , Células Intersticiais do Testículo , Masculino , Humanos , Células Intersticiais do Testículo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Homeostase , Testosterona
3.
Adv Sci (Weinh) ; 9(17): e2200626, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35435331

RESUMO

Spermatogonial stem cells regenerate and maintain spermatogenesis throughout life, making testis a good model for studying stem cell biology. The effects of chemotherapy on fertility have been well-documented previously. This study investigates how busulfan, an alkylating agent that is often used for chemotherapeutic purposes, affects male fertility. Specifically, the role of the TGR5 pathway is investigated on spermatogonia homeostasis using in vivo, in vitro, and pharmacological methods. In vivo studies are performed using wild-type and Tgr5-deficient mouse models. The results clearly show that Tgr5 deficiency can facilitate restoration of the spermatogonia homeostasis and allow faster resurgence of germ cell lineage after exposure to busulfan. TGR5 modulates the expression of key genes of undifferentiated spermatogonia such as Gfra1 and Fgfr2. At the molecular level, the present data highlight molecular mechanisms underlying the interactions among the TGR5, GLIS2, and TP53 pathways in spermatogonia associated with germ cell apoptosis following busulfan exposure. This study makes a significant contribution to the literature because it shows that TGR5 plays key role on undifferentiated germ cell homeostasis and that modulating the TGR5 signaling pathway could be used as a potential therapeutic tool for fertility disorders.


Assuntos
Bussulfano , Resistencia a Medicamentos Antineoplásicos , Fatores de Transcrição Kruppel-Like , Proteínas do Tecido Nervoso , Receptores Acoplados a Proteínas G , Proteína Supressora de Tumor p53 , Animais , Bussulfano/metabolismo , Bussulfano/farmacologia , Homeostase , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Proteínas do Tecido Nervoso/genética , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Espermatogônias/metabolismo , Proteína Supressora de Tumor p53/genética
4.
Cells ; 10(9)2021 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-34572051

RESUMO

Spermatogenesis is a process within the testis that leads to the production of spermatozoa. It is based on a population of spermatogonial stem cells, which have the capacity to self-renew and to differentiate throughout life to ensure the functions of reproduction are maintained. Male fertility disorders are responsible for half of the cases of infertility in couples worldwide. It is well known that cancer treatments are associated with reversible or irreversible fertility disorders. Busulfan (Bu) is an alkylating agent that significantly inhibits spermatogenesis. The present study relied on a combination of in vivo and in vitro approaches as well as RNAseq analysis to characterize the effects of Bu, in which mouse testes were used as a model. An in silico analysis revealed that many of the Bu-modulated genes are potentially regulated by the SIN3 Transcription Regulator Family Member A (SIN3A) and E2F Transcription Factor (E2F) families of transcription factors. The results demonstrate that the deregulated genes function in processes related to the cell cycle, DNA repair, and cell death mechanisms, including the Tumor Protein 53 (TP53) pathway. This reinforces the role of the TP53 signaling pathway as a major player in Bu effects. In addition, Bu altered the patterns of mRNA accumulation for various genes in undifferentiated spermatogonia. This work provides significant insight into the kinetics and impacts of busulfan, which could pave the way for developing strategies to minimize the impact of chemodrugs and, thus, could lead to germ cell lineage regeneration following anticancer treatments.


Assuntos
Bussulfano/farmacologia , Fertilidade/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Imunossupressores/farmacologia , Testículo/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA-Seq , Testículo/metabolismo
5.
Mol Cell Endocrinol ; 518: 110995, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32827571

RESUMO

The hypothalamic-pituitary axis exert a major control over endocrine and exocrine testicular functions. The hypothalamic-pituitary axis corresponds to a cascade with the Gonadotropin Releasing Hormone secreted by the hypothalamus, which stimulates the synthesis and the release of Luteinizing Hormone (LH) and Follicle Stimulating Hormone by the gonadotropic cells of the anterior pituitary. The LH signaling pathway controls the steroidogenic activity of the Leydig cells via the activation of the luteinizing hormone/choriogonadotropin receptor. In order to avoid a runaway system, sex steroids exert a negative feedback within hypothalamus and pituitary. Testicular steroidogenesis is locally controlled within Leydig cells. The present work reviews some local regulations of steroidogenesis within the Leydig cells focusing mainly on the roles of the Farnesoid-X-Receptor-alpha and its interactions with several orphan members of the nuclear receptor superfamily. Further studies are required to reinforce our knowledge of the regulation of testicular endocrine function, which is necessary to ensure a better understanding of fertility disorders and then proposed an adequate treatment of the diseases.


Assuntos
Hormônios Gonadais/metabolismo , Células Intersticiais do Testículo/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Células Endócrinas/metabolismo , Regulação da Expressão Gênica , Masculino , Camundongos , Hipófise/metabolismo , Ligação Proteica , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/fisiologia
6.
J Steroid Biochem Mol Biol ; 194: 105460, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31470110

RESUMO

The bile acid receptor Farnesoid-X-Receptor alpha (FXRα), a member of the nuclear receptor superfamily, is well known for its roles in the enterohepatic tract. In addition, FXRα regulates testicular physiology through the control of both endocrine and exocrine functions. The endocrine function of the Leydig cells is mainly controlled by the hypothalamo-pituitary axis viaLH/chorionic gonadotropin (CG). If FXRα was demonstrated to control the expression of the Lhcgr gene, encoding the LH receptor; the impact of the LH/CG signaling on the Fxrα expression has not been defined so far. Here, we demonstrate that hCG increases the Fxrα gene expression through the protein kinase-A signaling pathway. Fxrα is then involved in a negative feedback of steroid synthesis. These data improve our knowledge of the local control of the testicular steroidogenesis with the identification of the link between the hypothalamo-pituitary axis and the FXRα signaling pathway.


Assuntos
Gonadotropina Coriônica/farmacologia , Receptores Citoplasmáticos e Nucleares/genética , Testículo/efeitos dos fármacos , Animais , Linhagem Celular , Masculino , Camundongos Endogâmicos C57BL , Fosfoproteínas/genética , Progesterona/metabolismo , Receptores do LH/genética , Transdução de Sinais/efeitos dos fármacos , Testículo/metabolismo , Testosterona/sangue , Testosterona/metabolismo
7.
Stem Cell Reports ; 11(4): 944-958, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30245210

RESUMO

Several studies have reported an association between the farnesoid X receptor alpha (FXRα) and estrogenic signaling pathways. Fxrα could thus be involved in the reprotoxic effects of endocrine disruptors such as bisphenol-A (BPA). To test this hypothesis, mice were exposed to BPA and/or stigmasterol (S), an FXRα antagonist. Following the exposure to both molecules, wild-type animals showed impaired fertility and lower sperm cell production associated with the alteration of the establishment and maintenance of the undifferentiated germ cell pool. The crosstalk between BPA and FXRα is further supported by the lower impact of BPA in mice genetically ablated for Fxrα and the fact that BPA counteracted the effects of FXRα agonists. These effects might result from the downregulation of Fxrα expression following BPA exposure. BPA and S act additively in human testis. Our data demonstrate that FXRα activity modulates the impact of BPA on male gonads and on undifferentiated germ cell population.


Assuntos
Compostos Benzidrílicos/toxicidade , Diferenciação Celular , Células Germinativas/patologia , Homeostase , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Fenóis/toxicidade , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Adulto , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Feto/efeitos dos fármacos , Feto/patologia , Células Germinativas/efeitos dos fármacos , Células Germinativas/metabolismo , Homeostase/efeitos dos fármacos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estigmasterol/toxicidade
8.
Stem Cell Reports ; 9(1): 315-328, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28669602

RESUMO

Spermatogenesis is the process by which spermatozoa are generated from spermatogonia. This cell population is heterogeneous, with self-renewing spermatogonial stem cells (SSCs) and progenitor spermatogonia that will continue on a path of differentiation. Only SSCs have the ability to regenerate and sustain spermatogenesis. This makes the testis a good model to investigate stem cell biology. The Farnesoid X Receptor alpha (FXRα) was recently shown to be expressed in the testis. However, its global impact on germ cell homeostasis has not yet been studied. Here, using a phenotyping approach in Fxrα-/- mice, we describe unexpected roles of FXRα on germ cell physiology independent of its effects on somatic cells. FXRα helps establish and maintain an undifferentiated germ cell pool and in turn influences male fertility. FXRα regulates the expression of several pluripotency factors. Among these, in vitro approaches show that FXRα controls the expression of the pluripotency marker Lin28 in the germ cells.


Assuntos
Receptores Citoplasmáticos e Nucleares/metabolismo , Espermatogênese , Espermatozoides/citologia , Envelhecimento , Animais , Células Cultivadas , Feminino , Fertilidade , Deleção de Genes , Regulação da Expressão Gênica , Células Intersticiais do Testículo/citologia , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Reprodução , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Testículo/citologia , Testículo/metabolismo , Testículo/ultraestrutura
9.
Sci Rep ; 7: 42182, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28181583

RESUMO

Bile acids (BAs) are molecules with endocrine activities controlling several physiological functions such as immunity, glucose homeostasis, testicular physiology and male fertility. The role of the nuclear BA receptor FXRα in the control of BA homeostasis has been well characterized. The present study shows that testis synthetize BAs. We demonstrate that mice invalidated for the gene encoding FXRα have altered BA homeostasis in both liver and testis. In the absence of FXRα, BA exposure differently alters hepatic and testicular expression of genes involved in BA synthesis. Interestingly, Fxrα-/- males fed a diet supplemented with BAs show alterations of testicular physiology and sperm production. This phenotype was correlated with the altered testicular BA homeostasis and the production of intermediate metabolites of BAs which led to the modulation of CAR signaling pathways within the testis. The role of the CAR signaling pathways within testis was validated using specific CAR agonist (TCPOBOP) and inverse agonist (androstanol) that respectively inhibited or reproduced the phenotype observed in Fxrα-/- males fed BA-diet. These data open interesting perspectives to better define how BA homeostasis contributes to physiological or pathophysiological conditions via the modulation of CAR activity.


Assuntos
Ácidos e Sais Biliares/genética , Receptores Citoplasmáticos e Nucleares/genética , Reprodução/genética , Testículo/metabolismo , Androstanóis/farmacologia , Animais , Ácidos e Sais Biliares/biossíntese , Receptor Constitutivo de Androstano , Regulação da Expressão Gênica/efeitos dos fármacos , Homeostase/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Espermatozoides/metabolismo
10.
Cell Cycle ; 15(10): 1352-62, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27050906

RESUMO

The mammalian target of rapamycin (mTOR) plays essential roles in the regulation of growth-related processes such as protein synthesis, cell sizing and metabolism in both normal and pathological growing conditions. These functions of mTOR are thought to be largely a consequence of its cytoplasmic activity in regulating translation rate, but accumulating data highlight supplementary role(s) for this serine/threonine kinase within the nucleus. Indeed, the nuclear activities of mTOR are currently associated with the control of protein biosynthetic capacity through its ability to regulate the expression of gene products involved in the control of ribosomal biogenesis and proliferation. Using primary murine embryo fibroblasts (MEFs), we observed that cells with overactive mTOR signaling displayed higher abundance for the growth-associated Npm1 protein, in what represents a novel mechanism of Npm1 gene regulation. We show that Npm1 gene expression is dependent on mTOR as demonstrated by treatment of wild-type and Pten inactivated MEFs cultured with rapamycin or by transient transfections of small interfering RNA directed against mTOR. In accordance, the mTOR kinase localizes to the Npm1 promoter gene in vivo and it enhances the activity of a human NPM1-luciferase reporter gene providing an opportunity for direct control. Interestingly, rapamycin did not dislodge mTOR from the Npm1 promoter but rather strongly destabilized the Npm1 transcript by increasing its turnover. Using a prostate-specific Pten-deleted mouse model of cancer, Npm1 mRNA levels were found up-regulated and sensitive to rapamycin. Finally, we also showed that Npm1 is required to promote mTOR-dependent cell proliferation. We therefore proposed a model whereby mTOR is closely involved in the transcriptional and posttranscriptional regulation of Npm1 gene expression with implications in development and diseases including cancer.


Assuntos
Proteínas Nucleares/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Knockout , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Nucleofosmina , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Regiões Promotoras Genéticas , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Transplante Heterólogo , Regulação para Cima/efeitos dos fármacos
11.
Endocrinology ; 156(2): 660-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25426871

RESUMO

The small heterodimer partner (SHP, nuclear receptor subfamily 0, group B, member 2; NR0B2) is an atypical nuclear receptor known mainly for its role in bile acid homeostasis in the enterohepatic tract. We previously showed that NR0B2 controls testicular functions such as testosterone synthesis. Moreover, NR0B2 mediates the deleterious testicular effects of estrogenic endocrine disruptors leading to infertility. The endocrine homeostasis is essential for health, because it controls many physiological functions. This is supported by a large number of studies demonstrating that alterations of steroid activity lead to several kinds of diseases such as obesity and infertility. Within the testis, the functions of the Leydig cells are mainly controlled by the hypothalamo-pituitary axis via LH/chorionic gonadotropin (CG). Here, we show that LH/CG represses Nr0b2 expression through the protein kinase A-AMP protein kinase pathway. Moreover, using a transgenic mouse model invalidated for Nr0b2, we point out that NR0B2 mediates the repression of testosterone synthesis and subsequent germ cell apoptosis induced by exposure to anti-GnRH compound. Together, our data demonstrate a new link between hypothalamo-pituitary axis and NR0B2 in testicular androgen metabolism, making NR0B2 a major actor of testicular physiology in case of alteration of LH/CG levels.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiologia , Células Intersticiais do Testículo/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Testosterona/biossíntese , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Gonadotropina Coriônica , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos C57BL , Receptores LHRH/antagonistas & inibidores , Transdução de Sinais
12.
Expert Opin Ther Targets ; 18(12): 1367-76, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25263461

RESUMO

BACKGROUND: Liver physiology is sensitive to estrogens, which suggests that the liver might be a target of estrogenic endocrine disrupters (EED). However, the long-term consequences of neonatal exposure to EED on liver physiology have rarely been studied. The nuclear receptor small heterodimer partner (SHP) mediates the deleterious effects of neonatal exposure to diethylstilbestrol (DES) on male fertility. OBJECTIVES: As SHP is involved in liver homeostasis, we aimed to determine whether neonatal estrogenic exposure also affected adult liver physiology through SHP. Male mouse pups were exposed to DES in the first 5 days of life. RESULTS: DES exposure leads to alterations in the postnatal bile acid (BA) synthesis pathway. Neonatal DES-exposure affected adult liver BA metabolism and subsequently triglyceride (TG) homeostasis. The wild-type males neonatally exposed to DES exhibited increased liver weight and altered liver histology in the adult age. The use of deficient male mice revealed that SHP mediates the deleterious effects of DES treatment. These long-term effects of DES were associated with differently timed alterations in the expression of epigenetic factors. CONCLUSIONS: However, the molecular mechanisms by which neonatal exposure persist to affect the adult liver physiology remain to be defined. In conclusion, we demonstrate that neonatal DES exposure alters adult hepatic physiology in an SHP-dependent manner.


Assuntos
Dietilestilbestrol/toxicidade , Estrogênios não Esteroides/toxicidade , Fígado/efeitos dos fármacos , Fígado/fisiologia , Receptores Citoplasmáticos e Nucleares/deficiência , Fatores Etários , Animais , Animais Recém-Nascidos , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/fisiologia , Fígado/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout
13.
Biol Reprod ; 78(6): 968-75, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18276933

RESUMO

Human implantation involves invasion of the uterine wall and remodeling of uterine arteries by extravillous cytotrophoblasts. Defects in these early steps of placental development lead to poor placentation and are often associated with preeclampsia, a frequent complication of human pregnancy. One of the complex mechanisms controlling trophoblast invasion involves the activation of the liver X receptor beta (or NR1H2, more commonly known as LXRbeta) by oxysterols known as potent LXR activators. This activation of LXRbeta leads to a decrease of trophoblast invasion. The identification of new target genes of LXR in the placenta could aid in the understanding of their physiological roles in trophoblast invasion. In the present study, we show that the endoglin (ENG) gene is a direct target of the liver X receptor alpha (NR1H3, also known as LXRalpha). ENG, whose gene is highly expressed in syncytiotrophoblasts, is part of the transforming growth factor (TGF) receptor complex that binds several members of the TGFbeta superfamily. In the human placenta, ENG has been shown to be involved in the inhibition of trophoblast invasion. Treatment of human choriocarcinoma JAR cells with T0901317, a synthetic LXR-selective agonist, leads to a significant increase in ENG mRNA and protein levels. Using transfection and electrophoretic mobility shift assays, we demonstrate that LXR (as a heterodimer with the retinoid X receptor) is able to bind the ENG promoter on an LXR response element and mediates the activation of ENG gene expression by LXRalpha in JAR cells. This study suggests a novel mechanism by which LXR may regulate trophoblast invasion in pathological pregnancy such as preeclampsia.


Assuntos
Antígenos CD/genética , Antígenos CD/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Trofoblastos/metabolismo , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , DNA/genética , DNA/metabolismo , Primers do DNA/genética , Proteínas de Ligação a DNA/agonistas , Implantação do Embrião/genética , Implantação do Embrião/fisiologia , Endoglina , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hidrocarbonetos Fluorados , Ligantes , Receptores X do Fígado , Receptores Nucleares Órfãos , Pré-Eclâmpsia/etiologia , Gravidez , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores X de Retinoides/metabolismo , Sulfonamidas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...