Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 14: 1212716, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37720527

RESUMO

Cellular senescence is a response to a wide variety of stressors, including DNA damage, oncogene activation and physiologic aging, and pathologically accelerated senescence contributes to human disease, including diabetes mellitus. Indeed, recent work in this field has demonstrated a role for pancreatic ß-cell senescence in the pathogenesis of Type 1 Diabetes, Type 2 Diabetes and monogenic diabetes. Small molecule or genetic targeting of senescent ß-cells has shown promise as a novel therapeutic approach for preventing and treating diabetes. Despite these advances, major questions remain around the molecular mechanisms driving senescence in the ß-cell, identification of molecular markers that distinguish senescent from non-senescent ß-cell subpopulations, and translation of proof-of-concept therapies into novel treatments for diabetes in humans. Here, we summarize the current state of the field of ß-cell senescence, highlighting insights from mouse models as well as studies on human islets and ß-cells. We identify markers that have been used to detect ß-cell senescence to unify future research efforts in this field. We discuss emerging concepts of the natural history of senescence in ß-cells, heterogeneity of senescent ß-cells subpopulations, role of sex differences in senescent responses, and the consequences of senescence on integrated islet function and microenvironment. As a young and developing field, there remain many open research questions which need to be addressed to move senescence-targeted approaches towards clinical investigation.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Feminino , Masculino , Humanos , Animais , Camundongos , Diabetes Mellitus Tipo 2/terapia , Envelhecimento , Senescência Celular , Dano ao DNA
2.
J Clin Endocrinol Metab ; 106(1): 153-167, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-32961557

RESUMO

CONTEXT: Polycystic ovary syndrome (PCOS) is one of the leading causes of infertility, yet current diagnostic criteria are ineffective at identifying patients whose symptoms reside outside strict diagnostic criteria. As a result, PCOS is underdiagnosed and its etiology is poorly understood. OBJECTIVE: We aim to characterize the phenotypic spectrum of PCOS clinical features within and across racial and ethnic groups. METHODS: We developed a strictly defined PCOS algorithm (PCOSkeyword-strict) using the International Classification of Diseases, ninth and tenth revisions and keywords mined from clinical notes in electronic health records (EHRs) data. We then systematically relaxed the inclusion criteria to evaluate the change in epidemiological and genetic associations resulting in 3 subsequent algorithms (PCOScoded-broad, PCOScoded-strict, and PCOSkeyword-broad). We evaluated the performance of each phenotyping approach and characterized prominent clinical features observed in racially and ethnically diverse PCOS patients. RESULTS: The best performance came from the PCOScoded-strict algorithm, with a positive predictive value of 98%. Individuals classified as cases by this algorithm had significantly higher body mass index (BMI), insulin levels, free testosterone values, and genetic risk scores for PCOS, compared to controls. Median BMI was higher in African American females with PCOS compared to White and Hispanic females with PCOS. CONCLUSIONS: PCOS symptoms are observed across a severity spectrum that parallels the continuous genetic liability to PCOS in the general population. Racial and ethnic group differences exist in PCOS symptomology and metabolic health across different phenotyping strategies.


Assuntos
Algoritmos , Registros Eletrônicos de Saúde , Síndrome do Ovário Policístico , Adolescente , Adulto , Estudos de Casos e Controles , Interpretação Estatística de Dados , Mineração de Dados/métodos , Registros Eletrônicos de Saúde/estatística & dados numéricos , Etnicidade/genética , Etnicidade/estatística & dados numéricos , Feminino , Predisposição Genética para Doença/etnologia , Humanos , Herança Multifatorial , Fenótipo , Síndrome do Ovário Policístico/diagnóstico , Síndrome do Ovário Policístico/etnologia , Síndrome do Ovário Policístico/genética , Valor Preditivo dos Testes , Grupos Raciais/genética , Grupos Raciais/estatística & dados numéricos , Fatores de Risco , Tennessee/epidemiologia , Adulto Jovem
3.
Cell Rep ; 27(6): 1755-1768.e4, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-31067461

RESUMO

Preterm birth (PTB) is a syndrome with many origins. Among them, infection or inflammation are major risk factors for PTB; however, local defense mechanisms to mount anti-inflammatory responses against inflammation-induced PTB are poorly understood. Here, we show that endothelial TLR4 in the decidual bed is critical for sensing inflammation during pregnancy because mice with endothelial Tlr4 deletion are resistant to lipopolysaccharide (LPS)-induced PTB. Under inflammatory conditions, IL-6 is readily expressed in decidual endothelial cells with signal transducer and activator of transcription 3 (Stat3) phosphorylation in perivascular stromal cells, which then regulates expression of anti-inflammatory IL-10. Our observation that administration of an IL-10 neutralizing antibody predisposing mice to PTB shows IL-10's anti-inflammatory role to prevent PTB. We show that the integration of endothelial and perivascular stromal signaling can determine pregnancy outcomes. These findings highlight a role for endothelial TLR4 in inflammation-induced PTB and may offer a potential therapeutic target to prevent PTB.


Assuntos
Decídua/patologia , Células Endoteliais/metabolismo , Terapia de Alvo Molecular , Nascimento Prematuro/patologia , Nascimento Prematuro/prevenção & controle , Animais , Comunicação Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Feminino , Deleção de Genes , Inflamação/patologia , Interleucina-10/metabolismo , Interleucina-6/farmacologia , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos , Camundongos , NF-kappa B/metabolismo , Ovário/metabolismo , Gravidez , Fator de Transcrição STAT3/metabolismo , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/patologia , Receptor 4 Toll-Like/metabolismo
4.
Artigo em Inglês | MEDLINE | ID: mdl-30666294

RESUMO

Pheochromocytomas have been shown to impair glucose tolerance and, rarely, to precipitate overt diabetes mellitus. We report here a case of a large pheochromocytoma in a woman with a recent diagnosis of diabetes mellitus that proved difficult to control despite high-dose insulin therapy who had complete resolution of her hyperglycemia following adrenalectomy. Her dramatic presentation demonstrates the need to consider this etiology in patients with new-onset insulin resistance and hypertension.

5.
Am J Obstet Gynecol ; 217(5): 592.e1-592.e17, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28847437

RESUMO

BACKGROUND: Decidual senescence has been considered a mechanism of disease for spontaneous preterm labor in the absence of severe acute inflammation. Yet, signs of cellular senescence have also been observed in the chorioamniotic membranes from women who underwent the physiological process of labor at term. OBJECTIVE: We aimed to investigate whether, in the absence of acute histologic chorioamnionitis, the chorioamniotic membranes from women who underwent spontaneous preterm labor or labor at term exhibit signs of cellular senescence. STUDY DESIGN: Chorioamniotic membrane samples were collected from women who underwent spontaneous preterm labor or labor at term. Gestational age-matched nonlabor controls were also included. Senescence-associated genes/proteins were determined using reverse transcription quantitative polymerase chain reaction analysis (n = 7-9 each for array; n = 26-28 each for validation), enzyme-linked immunosorbent assays (n = 7-9 each), immunoblotting (n = 6-7 each), and immunohistochemistry (n = 7-8 each). Senescence-associated ß-galactosidase activity (n = 7-11 each) and telomere length (n = 15-22 each) were also evaluated. RESULTS: In the chorioamniotic membranes without acute histologic chorioamnionitis: (1) the expression profile of senescence-associated genes was different between the labor groups (term in labor and preterm in labor) and the nonlabor groups (term no labor and preterm no labor), yet there were differences between the term in labor and preterm in labor groups; (2) most of the differentially expressed genes among the groups were closely related to the tumor suppressor protein (TP53) pathway; (3) the expression of TP53 was down-regulated in the term in labor and preterm in labor groups compared to their nonlabor counterparts; (4) the expression of CDKN1A (gene coding for p21) was up-regulated in the term in labor and preterm in labor groups compared to their nonlabor counterparts; (5) the expression of the cyclin kinase CDK2 and cyclins CCNA2, CCNB1, and CCNE1 was down-regulated in the preterm in labor group compared to the preterm no labor group; (6) the concentration of TP53 was lower in the preterm in labor group than in the preterm no labor and term in labor groups; (7) the senescence-associated ß-galactosidase activity was greater in the preterm in labor group than in the preterm no labor and term in labor groups; (8) the concentration of phospho-S6 ribosomal protein was reduced in the term in labor group compared to its nonlabor counterpart, but no differences were observed between the preterm in labor and preterm no labor groups; and (9) no significant differences were observed in relative telomere length among the study groups (term no labor, term in labor, preterm no labor, and preterm in labor). CONCLUSION: In the absence of acute histologic chorioamnionitis, signs of cellular senescence are present in the chorioamniotic membranes from women who underwent spontaneous preterm labor compared to those who delivered preterm in the absence of labor. However, the chorioamniotic membranes from women who underwent spontaneous labor at term did not show consistent signs of cellular senescence in the absence of histologic chorioamnionitis. These results suggest that different pathways are implicated in the pathological and physiological processes of labor.


Assuntos
Âmnio/citologia , Senescência Celular/genética , Córion/citologia , Trabalho de Parto/genética , Trabalho de Parto Prematuro/genética , Adulto , Âmnio/metabolismo , Corioamnionite/patologia , Córion/metabolismo , Ciclina A2/genética , Ciclina B1/genética , Ciclina E/genética , Quinase 2 Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Feminino , Idade Gestacional , Humanos , Imuno-Histoquímica , Trabalho de Parto/metabolismo , Trabalho de Parto Prematuro/metabolismo , Proteínas Oncogênicas/genética , Fosfoproteínas/metabolismo , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína S6 Ribossômica/metabolismo , Transcriptoma , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , Adulto Jovem , beta-Galactosidase/metabolismo
6.
Sci Rep ; 6: 33023, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27620843

RESUMO

Here we demonstrate that conditional deletion of mouse uterine Trp53 (p53(d/d)), molecularly linked to mTORC1 activation and causally linked to premature uterine senescence and preterm birth, results in aberrant lipid signatures within the heterogeneous cell types of embryo implantation sites on day 8 of pregnancy. In situ nanospray desorption electrospray ionization mass spectrometry imaging (nano-DESI MSI) was used to characterize the molecular speciation of free fatty acids, monoacylglycerol species, unmodified and oxidized phosphatidylcholine (PC/Ox-PC), and diacylglycerol (DG) species within implantation sites of p53(d/d) mice and floxed littermates. Implantation sites from p53(d/d) mice exhibited distinct spatially resolved changes demonstrating accumulation of DG species, depletion of Ox-PC species, and increase in species with more unsaturated acyl chains, including arachidonic and docosahexaenoic acid. Understanding abnormal changes in the abundance and localization of individual lipid species early in the progression to premature birth is an important step toward discovering novel targets for treatments and diagnosis.


Assuntos
Implantação do Embrião/genética , Implantação do Embrião/fisiologia , Genes p53 , Metabolismo dos Lipídeos/genética , Útero/metabolismo , Animais , Diglicerídeos/metabolismo , Feminino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Knockout , Oxirredução , Fosfatidilcolinas/metabolismo , Gravidez , Espectrometria de Massas por Ionização por Electrospray
7.
J Clin Invest ; 126(8): 2941-54, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27454290

RESUMO

Inflammation and oxidative stress are known risk factors for preterm birth (PTB); however, the mechanisms and pathways that influence this condition are not fully described. Previously, we showed that mTORC1 signaling is increased in mice harboring a uterine-specific deletion of transformation-related protein 53 (p53d/d mice), which exhibit premature decidual senescence that triggers spontaneous and inflammation-induced PTB. Treatment with the mTORC1 inhibitor rapamycin reduced the incidence of PTB in the p53d/d mice. Decidual senescence with heightened mTORC1 signaling is also a signature of human PTB. Here, we have identified an underlying mechanism for PTB and a potential therapeutic strategy for treating the condition. Treatment of pregnant p53d/d mice with either the antidiabetic drug metformin or the antioxidant resveratrol activated AMPK signaling and inhibited mTORC1 signaling in decidual cells. Both metformin and resveratrol protected against spontaneous and inflammation-induced PTB in p53d/d females. Using multiple approaches, we determined that p53 interacts with sestrins to coordinate an inverse relationship between AMPK and mTORC1 signaling that determines parturition timing. This signature was also observed in human decidual cells. Together, these results reveal that p53-dependent coordination of AMPK and mTORC1 signaling controls parturition timing and suggest that metformin and resveratrol have therapeutic potential to prevent PTB.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Decídua/fisiologia , Complexos Multiproteicos/fisiologia , Proteínas Nucleares/fisiologia , Parto/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Antioxidantes/metabolismo , Sítios de Ligação , Senescência Celular , Feminino , Humanos , Inflamação , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Transgênicos , Estresse Oxidativo , Peroxidases , Gravidez , Nascimento Prematuro , Transdução de Sinais , Regulação para Cima , Útero/fisiologia
8.
Analyst ; 141(5): 1649-59, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26734689

RESUMO

Understanding how biological molecules are generated, metabolized and eliminated in living systems is important for interpreting processes such as immune response and disease pathology. While genomic and proteomic studies have provided vast amounts of information over the last several decades, interest in lipidomics has also grown due to improved analytical technologies revealing altered lipid metabolism in type 2 diabetes, cancer, and lipid storage disease. Mass spectrometry (MS) measurements are currently the dominant approach for characterizing the lipidome by providing detailed information on the spatial and temporal composition of lipids. However, interpreting lipids' biological roles is challenging due to the existence of numerous structural and stereoisomers (i.e. distinct acyl chain and double-bond positions), which are often unresolvable using present approaches. Here we show that combining liquid chromatography (LC) and structurally-based ion mobility spectrometry (IMS) measurement with MS analyses distinguishes lipid isomers and allows insight into biological and disease processes.


Assuntos
Cromatografia Líquida/métodos , Lipídeos/química , Espectrometria de Massas/métodos , Metabolômica/métodos , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Lipídeos/isolamento & purificação , Estereoisomerismo
9.
Cell Rep ; 8(2): 382-92, 2014 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-25043182

RESUMO

Embryo homing and implantation occur within a crypt (implantation chamber) at the antimesometrial (AM) pole along the uterus. The mechanism by which this is achieved is not known. Here, we show that villi-like epithelial projections from the main uterine lumen toward the AM pole at regularly spaced intervals that form crypts for embryo implantation were disrupted in mice with uterine loss or gain of function of Wnt5a, or loss of function of both Ror1 and Ror2. This disruption of Wnt5a-ROR signaling resulted in disorderly epithelial projections, crypt formation, embryo spacing, and impaired implantation. These early disturbances under abnormal Wnt5a-ROR signaling were reflected in adverse late pregnancy events, including defective decidualization and placentation, ultimately leading to compromised pregnancy outcomes. This study presents deeper insight regarding the formation of organized epithelial projections for crypt formation and embryo implantation for pregnancy success.


Assuntos
Decídua/metabolismo , Implantação do Embrião , Células Epiteliais/citologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Animais , Decídua/citologia , Decídua/fisiologia , Células Epiteliais/metabolismo , Feminino , Camundongos , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Proteínas Wnt/genética , Proteína Wnt-5a
10.
Mol Endocrinol ; 28(7): 1108-17, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24850415

RESUMO

Although cervical pregnancy and placenta previa, in which the embryo and placenta embed in or adjacent to the cervix, are life-threatening complications that result in massive bleeding and poor pregnancy outcomes in women, the incidence of these aberrant conditions is uncommon. We hypothesized that a local molecular mechanism is normally in place to prevent embryo implantation in the cervix. The ovarian hormones progesterone (P(4)) and estrogen differentially direct differentiation and proliferation of endometrial cells, which confers the receptive state for implantation: P(4) dominance causes differentiation of the luminal epithelium but increases stromal cell proliferation in preparation of the uterus for implantation. In search for the cause of cervical nonresponsiveness to implantation, we found that the statuses of cell proliferation and differentiation between the uterus and cervix during early pregnancy are remarkably disparate under identical endocrine milieu in both mice and humans. We also found that cervical levels of progesterone receptor (PR) protein are low compared with uterine levels during this period, and the low PR protein levels are attributed to elevated levels of microRNA(miR)-200a in the cervix. These changes were associated with up-regulation of the P(4)-metabolizing enzyme 20α-hydroxysteroid dehydrogenase (200α-HSD) and down-regulation of its transcriptional repressor signal transducer and activator of transcription 5 in the cervix. The results provide evidence that elevated levels of miR-200a lead to down-regulation of P(4)-PR signaling and up-regulation of (200α-HSD) in the cervix, rendering it nonresponsive to implantation. These findings may point toward not only the physiological but also the pathological basis of the cervical milieu in embryo implantation.


Assuntos
20-alfa-Hidroxiesteroide Desidrogenase/biossíntese , Colo do Útero/metabolismo , Implantação Tardia do Embrião/genética , MicroRNAs/genética , Progesterona/metabolismo , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Colo do Útero/patologia , Regulação para Baixo , Implantação do Embrião/genética , Endométrio/citologia , Endométrio/crescimento & desenvolvimento , Estrogênios/metabolismo , Feminino , Humanos , Antígeno Ki-67/metabolismo , Células MCF-7 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/biossíntese , Mifepristona/farmacologia , Gravidez , Progesterona/antagonistas & inibidores , Receptores de Progesterona/metabolismo , Fator de Transcrição STAT5/biossíntese , Células Estromais/citologia , Proteínas de Ligação a Tacrolimo/genética , Regulação para Cima , Útero/fisiologia
11.
J Clin Invest ; 123(9): 4063-75, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23979163

RESUMO

There are currently more than 15 million preterm births each year. We propose that gene-environment interaction is a major contributor to preterm birth. To address this experimentally, we generated a mouse model with uterine deletion of Trp53, which exhibits approximately 50% incidence of spontaneous preterm birth due to premature decidual senescence with increased mTORC1 activity and COX2 signaling. Here we provide evidence that this predisposition provoked preterm birth in 100% of females exposed to a mild inflammatory insult with LPS, revealing the high significance of gene-environment interactions in preterm birth. More intriguingly, preterm birth was rescued in LPS-treated Trp53-deficient mice when they were treated with a combination of rapamycin (mTORC1 inhibitor) and progesterone (P4), without adverse effects on maternal or fetal health. These results provide evidence for the cooperative contributions of two sites of action (decidua and ovary) toward preterm birth. Moreover, a similar signature of decidual senescence with increased mTORC1 and COX2 signaling was observed in women undergoing preterm birth. Collectively, our findings show that superimposition of inflammation on genetic predisposition results in high incidence of preterm birth and suggest that combined treatment with low doses of rapamycin and P4 may help reduce the incidence of preterm birth in high-risk women.


Assuntos
Decídua/metabolismo , Interação Gene-Ambiente , Nascimento Prematuro/prevenção & controle , 20-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Celecoxib , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Citocinas/metabolismo , Decídua/efeitos dos fármacos , Decídua/imunologia , Quimioterapia Combinada , Feminino , Expressão Gênica , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Lipopolissacarídeos/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Transgênicos , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/metabolismo , Ovário/efeitos dos fármacos , Ovário/imunologia , Ovário/patologia , Gravidez , Nascimento Prematuro/genética , Nascimento Prematuro/imunologia , Progesterona/farmacologia , Pirazóis/farmacologia , Receptores da Prolactina/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Sulfonamidas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
12.
Mol Hum Reprod ; 19(7): 463-72, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23475984

RESUMO

Heightened mammalian target of rapamycin complex 1 (mTORC1) activity by genetic deletion of its direct inhibitor, Tsc1, is associated with aberrant development and dysfunction of the female reproductive tract in mice. Here, we compared the phenotypes of mice with conditional deletion of Tsc1 in the female reproductive tract by either progesterone receptor (PR)-Cre (Tsc1(PR(d/d))), which inactivates Tsc1 in all major cell types in the uterus (epithelium, stroma and myometrium), or anti-Mullerian hormone type 2 receptor (Amhr2)-Cre (Tsc1(Amhr2(d/d))), which inactivates stromal and myometrial Tsc1. Tsc1(PR(d/d)) and Tsc1(Amhr2(d/d)) females are infertile resulting from oviductal hyperplasia, retention of embryos in the oviduct and implantation failure. In contrast to the appropriate embryonic development after fertilization seen in Tsc1(Amhr2(d/d)) females, embryo development was disrupted in Tsc1(PR(d/d)) females. In addition, uteri in Tsc1(PR(d/d)) and Tsc1(Amhr2(d/d)) females showed epithelial hyperplasia but not endometrial cancer. In conclusion, Tsc1(PR(d/d)) and Tsc1(Amhr2(d/d)) have overlapping yet distinct phenotypes in the context of compartment-specific deletion of Tsc1.


Assuntos
Complexos Multiproteicos/metabolismo , Oviductos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Útero/metabolismo , Animais , Estradiol/sangue , Feminino , Fertilização in vitro , Imuno-Histoquímica , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Mutantes , Complexos Multiproteicos/genética , Ovário/metabolismo , Progesterona/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina-Treonina Quinases TOR/genética , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
14.
Dev Cell ; 21(6): 1014-25, 2011 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-22100262

RESUMO

An effective bidirectional communication between an implantation-competent blastocyst and the receptive uterus is a prerequisite for mammalian reproduction. The blastocyst will implant only when this molecular cross-talk is established. Here we show that the muscle segment homeobox gene (Msh) family members Msx1 and Msx2, which are two highly conserved genes critical for epithelial-mesenchymal interactions during development, also play crucial roles in embryo implantation. Loss of Msx1/Msx2 expression correlates with altered uterine luminal epithelial cell polarity and affects E-cadherin/ß-catenin complex formation through the control of Wnt5a expression. Application of Wnt5a in vitro compromised blastocyst invasion and trophoblast outgrowth on cultured uterine epithelial cells. The finding that Msx1/Msx2 genes are critical for conferring uterine receptivity and readiness to implantation could have clinical significance, because compromised uterine receptivity is a major cause of pregnancy failure in IVF programs.


Assuntos
Implantação do Embrião/genética , Proteínas de Homeodomínio/genética , Fator de Transcrição MSX1/genética , Útero/metabolismo , Animais , Polaridade Celular/genética , Polaridade Celular/fisiologia , Implantação do Embrião/efeitos dos fármacos , Implantação do Embrião/fisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Fertilidade/genética , Fertilidade/fisiologia , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox , Proteínas de Homeodomínio/metabolismo , Fator Inibidor de Leucemia/administração & dosagem , Fator Inibidor de Leucemia/deficiência , Fator Inibidor de Leucemia/genética , Fator de Transcrição MSX1/deficiência , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Gravidez , Resultado da Gravidez , Útero/citologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Proteína Wnt-5a , beta Catenina/metabolismo
15.
Proc Natl Acad Sci U S A ; 108(44): 18073-8, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22025690

RESUMO

Although preterm delivery is a major global health issue, its causes and underlying mechanism remain elusive. Using mutant mice, mimicking aspects of human preterm birth, we show here that uterine decidual senescence early in pregnancy via heightened mammalian target of rapamycin complex 1 (mTORC1) signaling is a significant contributor of preterm birth and fetal death, and that these adverse phenotypes are rescued by a low dose of rapamycin, an inhibitor of mTORC1 signaling. This role of mTORC1 signaling in determining the timing of birth in mice may help us better understand the mechanism of the timing of birth in humans and develop new and improved strategies to combat the global problem of preterm birth.


Assuntos
Nascimento Prematuro , Proteínas/metabolismo , Transdução de Sinais , Útero/metabolismo , Animais , Western Blotting , Inibidor de Quinase Dependente de Ciclina p21/genética , Feminino , Morte Fetal , Imuno-Histoquímica , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos , Serina-Treonina Quinases TOR , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA