Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Front Immunol ; 14: 1276979, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38022561

RESUMEN

Pregnancy success is dependent on the establishment of maternal tolerance during the preimplantation period. The immunosuppressive function of regulatory T cells is critical to limit inflammation arising from implantation of the semi-allogeneic blastocyst. Insufficient maternal immune adaptations to pregnancy have been frequently associated with cases of female infertility and recurrent implantation failure. The role of Nodal, a secreted morphogen of the TGFß superfamily, was recently implicated during murine pregnancy as its conditional deletion (NodalΔ/Δ) in the female reproductive tract resulted in severe subfertility. Here, it was determined that despite normal preimplantation processes and healthy, viable embryos, NodalΔ/Δ females had a 50% implantation failure rate compared to NodalloxP/loxP controls. Prior to implantation, the expression of inflammatory cytokines MCP-1, G-CSF, IFN-γ and IL-10 was dysregulated in the NodalΔ/Δ uterus. Further analysis of the preimplantation leukocyte populations in NodalΔ/Δ uteri showed an overabundance of infiltrating, pro-inflammatory CD11bhigh Ly6C+ macrophages coupled with the absence of CD4+ FOXP3+ regulatory T cells. Therefore, it is proposed that uterine Nodal expression during the preimplantation period has a novel role in the establishment of maternal immunotolerance, and its dysregulation should be considered as a potential contributor to cases of female infertility and recurrent implantation failure.


Asunto(s)
Infertilidad Femenina , Proteína Nodal , Linfocitos T Reguladores , Animales , Femenino , Ratones , Embarazo , Implantación del Embrión , Factores de Transcripción Forkhead , Infertilidad Femenina/genética , Útero , Proteína Nodal/genética
2.
Reprod Biol Endocrinol ; 21(1): 85, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37715251

RESUMEN

Adhesion of the implanting blastocyst involves the interaction between integrin proteins expressed by trophoblast cells and components present in the basement membrane of the endometrial luminal epithelium. Although several factors regulating integrins and their adhesion to fibronectin are already known, we showed that Wnt signaling is involved in the regulation of blastocyst adhesion through the trafficking of integrins expressed by trophoblast cells. Localization of Itgα5ß1 by immunofluorescence and FN-binding assays were conducted on peri-implantation blastocysts treated with either Wnt5a or Wnt7a proteins. Both Wnt5a and Wnt7a induced a translocation of Itgα5ß1 at the surface of the blastocyst and an increase in FN-binding activity. We further demonstrated that uterine fluid is capable of inducing integrin translocation and this activity can be specifically inhibited by the Wnt inhibitor sFRP2. To identify the Wnt signaling pathway involved in this activity, blastocysts were incubated with inhibitors of either p38MAPK, PI3K pathway or CamKII prior to the addition of Wnts. Whereas inhibition of p38MAPK and PI3K had not effect, inhibition of CamKII reduced FN-binding activity induced by Wnts. Finally, we demonstrated that inhibition of Wnts by sFRP2 reduced the binding efficiency of the blastocyst to uterine epithelial cells. Our findings provide new insight into the mechanism that regulates integrin trafficking and FN-binding activity and identifies Wnts as a key player in blastocyst attachment to the uterine epithelium.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Fibronectinas , Animales , Ratones , Femenino , Fosfatidilinositol 3-Quinasas , Blastocisto , Vía de Señalización Wnt , Integrinas
4.
Placenta ; 107: 13-23, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33730615

RESUMEN

INTRODUCTION: The growth and survival of the mammalian fetus is highly dependent on the placenta. Several research groups have demonstrated the involvement of different transforming growth factor-beta (TGFß) superfamily members and their related receptors in placentation. Cripto is a member of the epidermal growth factor-Cripto1/FRL1/Cryptic protein family and plays a critical role in embryonic development, stem cell maintenance and tumor progression through TGFß-dependent and independent pathways. Several studies have suggested that Cripto may also have a role in female reproduction and pregnancy maintenance, but its specific role remains elusive. METHODS: We used a conditional knockout mouse model in which Cripto is deleted from the uterus using a loxP-Cre system. Cripto cKO females were mated with wildtype males and dissections were performed at different timepoints during pregnancy for assessment of the number and size of the implantation sites, resorption sites, fetal weight and placental development. Histology, IF staining and quantitative PCR were employed to analyze the placentation process. RESULTS: We found that loss of maternal Cripto results in defective placentation, decreased vascularization within the placental labyrinth and leads to intrauterine growth restriction and fetal death. We further demonstrated that components of the VEGF and Notch signaling pathways are downregulated in Cripto cKO decidua and placenta potentially contributing to defects in the development of the vasculature at maternal-fetal interface. DISCUSSION: These findings demonstrate that maternal Cripto is involved in the maternal-fetal communications required for proper development of the placenta and placental vasculature.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Placenta/irrigación sanguínea , Placentación/fisiología , Animales , Factor de Crecimiento Epidérmico/genética , Femenino , Fertilidad/fisiología , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Placenta/metabolismo , Embarazo , Trofoblastos/metabolismo , Útero/metabolismo
5.
Biol Reprod ; 104(5): 1045-1057, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33590845

RESUMEN

Cripto encodes for a cell surface receptor whose role in embryonic development and stem cell maintenance has been studied. Cripto mRNA and protein have been detected in the human uterus at all stages of the menstrual cycle. To date, there is not much known about Cripto's role in female reproduction. As Cripto null Knockout (KO) is embryonic lethal, we created a conditional KO (cKO) mouse model in which Cripto is deleted only in the reproductive tissues using a Cre-loxP system. Pregnancy rate and number of pups per litter were evaluated as general fertility indices. We observed a significant decrease in pregnancy rate and litter size with loss of uterine Cripto indicating that Cripto cKO females are subfertile. We showed that although the preimplantation period is normal in Cripto cKO females, 20% of cKO females fail to establish pregnancy and an additional 20% of females undergo full litter loss after implantation between day 5.5 postcoitum (d5.5pc) and d8.5pc. We showed that subfertility caused by loss of uterine Cripto is due to defects in uterine decidualization, remodeling, and luminal closure and is accompanied by significant downregulation of Bmp2, Wnt4 and several components of Notch signaling pathway which all are known to be important factors in uterine remodeling and decidualization. Our study demonstrates that Cripto is expressed in the uterus during critical stages of early pregnancy and its deletion results in subfertility due to implantation failure, impaired peri-implantation uterine remodeling and impaired uterine decidualization.


Asunto(s)
Decidua/metabolismo , Implantación del Embrión/genética , Factor de Crecimiento Epidérmico/genética , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/genética , Útero/metabolismo , Animales , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Glicoproteínas de Membrana/metabolismo , Ratones , Proteínas de Neoplasias/metabolismo
6.
Angiogenesis ; 23(3): 443-458, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32385775

RESUMEN

During the initiation of pregnancy, the vasculature of the implantation site expands rapidly, yet little is known about this process or its role in fertility. Here, we report that endothelial-specific deletion of a disintegrin and metalloprotease 10 (ADAM10), an essential regulator of Notch signaling, results in severe subfertility in mice. We found that implantation sites develop until 5.5 days post conception (dpc) but are resorbed by 6.5 dpc in A10ΔEC mice. Analysis of the mutant implantation sites showed impaired decidualization and abnormal vascular patterning compared to controls. Moreover, RNA-seq analysis revealed changes in endothelial cell marker expression consistent with defective ADAM10/Notch signaling in samples from A10ΔEC mice, suggesting that this signaling pathways is essential for the physiological function of endometrial endothelial cells during early pregnancy. Our findings raise the possibility that impaired endothelial cell function could be a cause for repeated pregnancy loss (RPL) and infertility in humans.


Asunto(s)
Proteína ADAM10/deficiencia , Secretasas de la Proteína Precursora del Amiloide/deficiencia , Decidua/metabolismo , Fertilidad , Eliminación de Gen , Proteínas de la Membrana/deficiencia , Receptores Notch/metabolismo , Transducción de Señal , Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Femenino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Embarazo , Receptores Notch/genética
7.
Biol Reprod ; 102(6): 1340-1350, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32219302

RESUMEN

Preterm birth remains the major cause of perinatal mortality and morbidity worldwide, affecting up to 12% of pregnancies and accounting for ~75% of neonatal deaths. However, the mechanisms and causes that underlie it are still largely unknown. One of the major causes of preterm birth is infection or inflammation within the maternal-fetal interface. Our lab has previously shown that a uterine specific deletion of Nodal results in mutant females delivering 2 days prior to term demonstrating an important role for this factor in the maintenance of pregnancy. Here, we have addressed the function of Nodal in the uterus during pregnancy. We demonstrate that Nodal heterozygous mice have an increase in basal levels of pro-inflammatory cytokines IL-1ß, IL-6, IL-12p, TNF-α, and IFN-γ as well as an increase in the number of macrophages in response to the inflammatory agent, lipopolysaccharide (LPS). Using bone marrow-derived macrophages, we demonstrated that pretreatment with recombinant Nodal reduces pro-inflammatory gene expression when these cells are challenged with LPS. Our results demonstrate that Nodal is required to maintain the uterine environment in an anti-inflammatory state by preventing proinflammatory cytokine expression.


Asunto(s)
Citocinas/metabolismo , Proteína Nodal/metabolismo , Útero/fisiología , Animales , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Inflamación/metabolismo , Células Asesinas Naturales , Macrófagos/fisiología , Ratones , Proteína Nodal/genética , Fosforilación , Placenta/metabolismo , Embarazo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
8.
J Perinatol ; 38(5): 482-488, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29453435

RESUMEN

OBJECTIVE: NODAL has been implicated in timing of parturition and immune regulation. We investigated the relationship between NODAL polymorphisms, infection/inflammation, and preterm birth. STUDY DESIGN: For this secondary analysis, 613 women (189 preterm and 424 term) from the Montreal Prematurity Study were genotyped for NODAL polymorphisms and assessed for bacterial vaginosis and placental inflammation. RESULT: NODAL polymorphisms were not associated with preterm birth. However, the rs2231947(C>T) variant allele was associated with increased risk for preterm birth among women with bacterial vaginosis (odds ratio: 2.76, 95% confidence interval: 1.12-6.85). Among women without placental inflammation, the rs1904589(A>G) variant allele was associated with increased risk of preterm birth (odds ratio: 1.31, 95% confidence interval: 1.02-1.70). Among women with placental inflammation, the rs10999338(C>T) variant allele was associated with reduced risk of preterm birth (odds ratio: 0.50, 95% confidence interval: 0.29-0.87). CONCLUSION: The effect of NODAL polymorphisms on preterm birth depends on maternal infection/inflammation status.


Asunto(s)
Interacción Gen-Ambiente , Proteína Nodal/genética , Trabajo de Parto Prematuro/genética , Complicaciones Infecciosas del Embarazo/diagnóstico , Vaginosis Bacteriana/genética , Adolescente , Adulto , Alelos , Estudios de Casos y Controles , Femenino , Humanos , Recién Nacido de Bajo Peso , Recién Nacido , Inflamación , Modelos Logísticos , Masculino , Oportunidad Relativa , Polimorfismo Genético , Embarazo , Estudios Prospectivos , Vaginosis Bacteriana/complicaciones , Vaginosis Bacteriana/diagnóstico , Adulto Joven
9.
Reproduction ; 155(1): 93-102, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29066530

RESUMEN

Six members of the Wnt family are expressed in the female reproductive tract. Their collective function ensures proper development of the uterus, preparing it for pregnancy during adulthood. Here, we take advantage of the fact that Porcn, a prerequisite for all Wnt secretion, is located on the X chromosome, to generate females that were mosaic for Porcn throughout the reproductive tract. Porcnflox/+ females were mated with progesterone receptor (Pgr)-Cre males (PgrCre/+ ) to generate females that were heterozygous for Porcupine in all tissues of the female reproductive tract, resulting in mosaicism due to random X-inactivation. We demonstrated that Porcn mosaic females are extremely subfertile and exhibit a large spectrum of phenotypes ranging from morphologically normal uteri to uteri with extremely enlarged cystic glands. Decreased fertility in Porcupine mosaic females was not associated with phenotype severity and was observed regardless of whether or not cystic glands were enlarged. By crossing-in a GFP reporter on the wild-type X chromosome, we were able to correlate endometrial gland hyperplasia with a mostly Porcupine mutant stroma, demonstrating the role of stromal Wnts in the regulation of endometrial gland proliferation. Finally, we demonstrated that fertility issues within mosaic females were due to a reduced response to estrogen and to abnormal Tcf/Lef signaling across the mesometrial-anti-mesometrial axis during the window of implantation.


Asunto(s)
Aciltransferasas/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Infertilidad/etiología , Proteínas de la Membrana/fisiología , Organogénesis , Células del Estroma/patología , Útero/patología , Proteínas Wnt/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Implantación del Embrión , Estrógenos/farmacología , Femenino , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Infertilidad/metabolismo , Infertilidad/patología , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Masculino , Ratones , Ratones Noqueados , Embarazo , Reproducción , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Útero/efectos de los fármacos , Útero/metabolismo , Proteínas Wnt/genética
10.
Biol Reprod ; 97(5): 688-697, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29036275

RESUMEN

The secretion of mammalian Wnt ligands within the cell is dependent on the activity of Porcupine, a gene located on the X-chromosome that encodes for a membrane-bound O-acyl transferase. Here, we report that postnatal ablation of Porcupine in the uterine luminal epithelium alone results in the decrease in endometrial gland number. Despite having uterine glands, mutant females are completely infertile. Epithelial ablation of Porcupine causes defects in timely apposition of the lumen, along with failure to respond to artificial decidual induction. Interestingly, progesterone supplementation was able to rescue the initiation of decidualization, but the decidua was not maintained and subsequently resorbed. Transcriptome analysis demonstrated that deletion of Porcupine in the epithelium resulted in the stromal dysregulation of members of the Wnt signaling pathway (Lef1, Wnt4, and Wnt16), dysregulation of receptors and ligands in the Notch signaling pathway (Notch1, Notch4, and Dll4) as well as Hoxa10. Our results demonstrate the crucial requirement of Wnt signaling in the epithelium for fertility and demonstrate that epithelial Wnts regulate stromal Wnt gene expression as well as regulating the expression of essential signaling factors and effectors required for successful embryo implantation.


Asunto(s)
Aciltransferasas/metabolismo , Fertilidad/fisiología , Proteínas de la Membrana/metabolismo , Útero/metabolismo , Proteínas Wnt/metabolismo , Aciltransferasas/genética , Animales , Epitelio/metabolismo , Femenino , Fertilidad/genética , Regulación de la Expresión Génica/fisiología , Masculino , Proteínas de la Membrana/genética , Ratones , Mutación , Transducción de Señal/fisiología , Proteínas Wnt/genética
11.
Dev Biol ; 422(1): 58-69, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27965056

RESUMEN

Wnt signaling has been shown to be important in orchestrating proper development of the female reproductive tract. In the uterus, six members of the Wnt family are expressed in the neonatal endometrium and deletion of individual Wnt genes often leads to similar phenotypes, suggesting an interaction of these genes in uterine development and function. Furthermore, Wnts may have complementary functions, which could mask the identification of their individual functional role in single gene deletions. To circumvent this issue, we have generated a deletion of the Porcupine homolog within the female reproductive tract using progesterone receptor-Cre mice (PgrCre/+); preventing Wnt secretion from the producing cells. We show that Porcupine-dependent Wnt signaling, unlike previously reported, is dispensable for postnatal gland formation but is required for post-pubertal gland maintenance as well as for stromal cell proliferation. Furthermore, our results demonstrate that WNT7a is sufficient to restore post-pubertal endometrial gland formation. Although WNT5a did not restore gland formation, it rescued stromal cell proliferation; up-regulating several secreted factors including Fgf10 and Ihh. Our results further elucidate the roles of Wnt signaling in uterine development and function as well as provide an ideal system to address individual Wnt functions in the uterus.


Asunto(s)
Endometrio/citología , Proteínas de la Membrana/fisiología , Proteínas Wnt/fisiología , Vía de Señalización Wnt/fisiología , Proteína Wnt-5a/fisiología , Aciltransferasas , Animales , Comunicación Celular , Proliferación Celular , Femenino , Ratones , Células del Estroma/fisiología
12.
Front Genet ; 4: 170, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23986775

RESUMEN

Nodal, a secreted signaling protein from the transforming growth factor beta (TGF-ß)-super family plays a vital role during early embryonic development. Recently, it was found that maternal decidua-specific Nodal knockout mice show intrauterine growth restriction (IUGR) and preterm birth. The chromosomal location of NODAL is in the same linkage area as the placental (fetal) pre-eclampsia (PE) susceptibility gene STOX1, which is associated with the familial form of early-onset, IUGR-complicated PE. As the STOX1 linkage was originally identified in women being born from a pre-eclamptic pregnancy as well as suffering from PE themselves, the linkage could in part be caused by NODAL, which is why the potential maternal-fetal interaction between STOX1 and NODAL was investigated. In the PE families with the STOX1 susceptibility allele carried by the children born from pre-eclamptic pregnancies, it was found that the pre-eclamptic mothers themselves all carried the NODAL H165R SNP, which causes a 50% reduced activity. Surprisingly, in decidua-specific Nodal knockout mice the fetal placenta showed up-regulation of STOX1 and NODAL expression. Conditioned media of human first trimester decidua and a human endometrial stromal cell line (T-HESC) treated with siRNAs against NODAL or carrying the H165R SNP were also able to induce NODAL and STOX1 expression when added to SGHPL-5 first trimester extravillous trophoblast cells. Finally, a human TGF-ß/BMP signaling pathway PCR-array on decidua and the T-HESC cell line with Nodal knockdown revealed upregulation of Activin-A, which was confirmed in conditioned media by ELISA. We show that maternal decidua Nodal knockdown gives upregulation of NODAL and STOX1 mRNA expression in fetal extravillous trophoblast cells, potentially via upregulation of Activin-A in the maternal decidua. As both Activin-A and Nodal have been implicated in PE, being increased in serum of pre-eclamptic women and upregulated in pre-eclamptic placentas respectively, this interaction at the maternal-fetal interface might play a substantial role in the development of PE.

13.
Reproduction ; 145(2): R55-64, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23221013

RESUMEN

Successful mammalian reproduction is dependent on a receptive and nurturing uterine environment. In order to establish pregnancy in humans, the uterus must i) be adequately prepared to receive the blastocyst, ii) engage in a coordinated molecular dialog with the embryo to facilitate implantation, and iii) undergo endometrial decidualization. Although numerous factors have been implicated in these essential processes, the precise network of molecular interactions that govern receptivity, embryo implantation, and decidualization remain unclear. NODAL, a morphogen in the transforming growth factor ß superfamily, is well known for its critical functions during embryogenesis; however, recent studies have demonstrated an emerging role for NODAL signaling during early mammalian reproduction. Here, we review the established data and a recent wave of new studies implicating NODAL signaling components in uterine cycling, embryo implantation, and endometrial decidualization in humans and mice.


Asunto(s)
Implantación del Embrión/genética , Proteína Nodal/fisiología , Embarazo/genética , Animales , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Femenino , Humanos , Ratones , Proteína Nodal/genética , Proteína Nodal/metabolismo , Embarazo/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Especificidad de la Especie
14.
Biol Reprod ; 86(6): 194, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22378764

RESUMEN

Preterm birth is the single leading cause of perinatal mortality in developed countries, affecting approximately 12% of pregnancies and accounting for 75% of neonatal loss in the United States. Despite the prevalence and severity of premature delivery, the causes and mechanisms that underlie spontaneous and idiopathic preterm birth remain unknown. Our inability to elucidate these fundamental causes has been attributed to a poor understanding of the signaling pathways associated with the premature induction of parturition and a lack of suitable animal models available for preterm birth research. In this study, we describe the generation and analysis of a novel conditional knockout of the transforming growth factor beta (TGFB) superfamily member, Nodal, from the maternal reproductive tract of mice. Strikingly, uterine Nodal knockout females exhibited a severe malformation of the maternal decidua basalis during placentation, leading to significant intrauterine growth restriction, and ultimately preterm birth and fetal loss on Day 17.5 of gestation. Using several approaches, we characterized aberrant placental development and demonstrated that reduced proliferation combined with increased apoptosis resulted in a diminished decidua basalis and compromised maternal-fetal interface. Last, we evaluated various components of the established parturition cascade and determined that preterm birth derived from the maternal Nodal knockout occurs prior to PTGS2 (COX-2) upregulation at the placental interface. Taken together, the results presented in this study highlight an in vivo role for maternal NODAL during placentation, present an interesting link between disrupted decidua basalis formation and premature parturition, and describe a potentially valuable model toward elucidating the complex processes that underlie preterm birth.


Asunto(s)
Proteína Nodal/metabolismo , Parto/metabolismo , Placentación , Nacimiento Prematuro/metabolismo , Útero/metabolismo , Animales , Apoptosis , Proliferación Celular , Femenino , Fertilidad , Muerte Fetal , Retardo del Crecimiento Fetal/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placenta/anomalías , Embarazo
15.
Eur J Neurosci ; 33(8): 1366-75, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21375603

RESUMEN

Neurons and glia in the central nervous system originate from neural stem and progenitor cells that reside in the ventricular zones. Here we examine the role of ß-catenin in neural stem cell (NSC) regulation in mouse embryos lacking ß-catenin specifically in the brain germinal zone. An in vitro clonal neurosphere assay was performed in order to ascertain the status of the NSC population. Intact neurospheres did not form from ß-catenin-null cells due to a loss of cell adhesion and the number of expanded cells was reduced. Rescue of ß-catenin expression restored adhesion and revealed that the number of NSCs increased in the knockout population. Using a clonal colony-forming assay, which confines precursor cells within a solid collagen matrix, we show that the number of NSCs in the hippocampus is unchanged although the ß-catenin knockout striatum actually contains a larger proportion of NSCs. However, these colonies were smaller than those of control cells, due to increased apoptosis in the progenitor population. Furthermore, ß-catenin knockout NSCs also retained multipotentiality as shown by their ability to clonally differentiate into neurons and glia. The effects on neural precursor cells were not due to loss of downstream T-cell factor signaling, as this pathway is not active in vivo in regions of the embryonic brain where NSCs and progenitor cells reside, nor is it active in vitro in NSC colonies. These data reveal that ß-catenin is not required for the maintenance or differentiation of NSCs, but is required for the adhesion and survival of neural progenitor cells.


Asunto(s)
Muerte Celular/fisiología , Células-Madre Neurales/fisiología , Células Madre/fisiología , beta Catenina/metabolismo , Animales , Adhesión Celular , Diferenciación Celular/fisiología , Supervivencia Celular , Células Cultivadas , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/citología , Neuroglía/citología , Neuroglía/fisiología , Transducción de Señal/fisiología , Células Madre/citología , beta Catenina/genética
16.
Biol Reprod ; 84(6): 1103-10, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21270430

RESUMEN

Nodal, a transforming growth factor beta (TGFB) superfamily member, plays a critical role during early embryonic development. Recently, components of the Nodal signaling pathway were characterized in the human uterus and implicated in the tissue remodeling events during menstruation. Furthermore, the Nodal inhibitor, Lefty, was identified in the mouse endometrium during pregnancy, and its overexpression led to implantation failure. Nonetheless, the precise function and mechanism of Nodal signaling during pregnancy remains unclear. In order to elucidate the potential roles Nodal plays in these processes, we have generated a detailed profile of maternal Nodal expression in the mouse uterus throughout pregnancy. NODAL, although undetectable during the nonpregnant estrus cycle, was localized throughout the glandular epithelium of the endometrium during the peri-implantation period. Interestingly, Nodal expression generated a banding pattern along the proximal-distal axis of the uterine horn on Day 4.5 that directly correlated with blastocyst implantation. Embryo transfer experiments indicate the embryo regulates Nodal expression in the uterus and directs its expression at the time of implantation, restricting NODAL to the sites between implantation crypts. During the later stages of pregnancy, Nodal exhibits a dynamic expression profile that suggests a role in regulating the endometrial response to decidualization and associated trophoblast invasion.


Asunto(s)
Implantación del Embrión/fisiología , Regulación de la Expresión Génica/fisiología , Proteína Nodal/metabolismo , Útero/metabolismo , Animales , Transferencia de Embrión , Femenino , Perfilación de la Expresión Génica , Genes Transgénicos Suicidas , Factores de Determinación Derecha-Izquierda/genética , Factores de Determinación Derecha-Izquierda/metabolismo , Ratones , Proteína Nodal/genética , Embarazo
17.
Mol Cell Biol ; 31(1): 163-78, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21041481

RESUMEN

Specification and early patterning of the vertebrate heart are dependent on both canonical and noncanonical wingless (Wnt) signal pathways. However, the impact of each Wnt pathway on the later stages of myocardial development and differentiation remains controversial. Here, we report that the components of each Wnt signal conduit are expressed in the developing and postnatal heart, yet canonical/ß-catenin activity is restricted to nonmyocardial regions. Subsequently, we observed that noncanonical Wnt (Wnt11) enhanced myocyte differentiation while preventing stabilization of the ß-catenin protein, suggesting active repression of canonical Wnt signals. Wnt11 stimulation was synonymous with activation of a caspase 3 signal cascade, while inhibition of caspase activity led to accumulation of ß-catenin and a dramatic reduction in myocyte differentiation. Taken together, these results suggest that noncanonical Wnt signals promote myocyte maturation through caspase-mediated inhibition of ß-catenin activity.


Asunto(s)
Caspasas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas Wnt/metabolismo , Animales , Secuencia de Bases , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Sondas de ADN/genética , Femenino , Corazón Fetal/citología , Corazón Fetal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ratones , Ratones Transgénicos , Embarazo , Estabilidad Proteica , Transducción de Señal , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/genética , beta Catenina/antagonistas & inhibidores , beta Catenina/metabolismo
18.
BMC Dev Biol ; 10: 121, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21176145

RESUMEN

BACKGROUND: Understanding the dynamic cellular behaviors and underlying molecular mechanisms that drive morphogenesis is an ongoing challenge in biology. Live imaging provides the necessary methodology to unravel the synergistic and stereotypical cell and molecular events that shape the embryo. Genetically-encoded reporters represent an essential tool for live imaging. Reporter strains can be engineered by placing cis-regulatory elements of interest to direct the expression of a desired reporter gene. In the case of canonical Wnt signaling, also referred to as Wnt/ß-catenin signaling, since the downstream transcriptional response is well understood, reporters can be designed that reflect sites of active Wnt signaling, as opposed to sites of gene transcription, as is the case with many fluorescent reporters. However, even though several transgenic Wnt/ß-catenin reporter strains have been generated, to date, none provides the single-cell resolution favored for live imaging studies. RESULTS: We have placed six copies of a TCF/Lef responsive element and an hsp68 minimal promoter in front of a fluorescent protein fusion comprising human histone H2B to GFP and used it to generate a strain of mice that would report Wnt/ß-catenin signaling activity. Characterization of developmental and adult stages of the resulting TCF/Lef:H2B-GFP strain revealed discrete and specific expression of the transgene at previously characterized sites of Wnt/ß-catenin signaling. In support of the increased sensitivity of the TCF/Lef:H2B-GFP reporter, additional sites of Wnt/ß-catenin signaling not documented with other reporters but identified through genetic and embryological analysis were observed. Furthermore, the sub-cellular localization of the reporter minimized reporter perdurance, and allowed visualization and tracking of individual cells within a cohort, so facilitating the detailed analysis of cell behaviors and signaling activity during morphogenesis. CONCLUSION: By combining the Wnt activity read-out efficiency of multimerized TCF/Lef DNA binding sites, together with the high-resolution imaging afforded by subcellularly-localized fluorescent fusion proteins such as H2B-GFP, we have created a mouse transgenic line that faithfully recapitulates Wnt signaling activity at single-cell resolution. The TCF/Lef:H2B-GFP reporter represents a unique tool for live imaging the in vivo processes triggered by Wnt/ß-catenin signaling, and thus should help the formulation of a high-resolution understanding of the serial events that define the morphogenetic process regulated by this signaling pathway.


Asunto(s)
Ratones/embriología , Transducción de Señal , Análisis de la Célula Individual/métodos , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Endodermo/metabolismo , Gastrulación , Proteínas Fluorescentes Verdes/metabolismo , Ratones Transgénicos , Línea Primitiva/metabolismo , Factores de Transcripción TCF/metabolismo
19.
Biol Reprod ; 83(1): 52-62, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20130269

RESUMEN

Mammalian mitochondrial DNA (mtDNA) is a small, maternally inherited genome that codes for 13 essential proteins in the respiratory chain. Mature oocytes contain more than 150 000 copies of mtDNA, at least an order of magnitude greater than the number in most somatic cells, but sperm contain only approximately 100 copies. Mitochondrial oxidative phosphorylation has been suggested to be an important determinant of oocyte quality and sperm motility; however, the functional significance of the high mtDNA copy number in oocytes, and of the low copy number in sperm, remains unclear. To investigate the effects of mtDNA copy number on fertility, we genetically manipulated mtDNA copy number in the mouse by deleting one copy of Tfam, an essential component of the mitochondrial nucleoid, at different stages of germline development. We show that males can tolerate at least a threefold reduction in mtDNA copy number in their sperm without impaired fertility, and in fact, they preferentially transmit a deleted Tfam allele. Surprisingly, oocytes with as few as 4000 copies of mtDNA can be fertilized and progress normally through preimplantation development to the blastocyst stage. The mature oocyte, however, has a critical postimplantation developmental threshold of 40 000-50 000 copies of mtDNA in the mature oocyte. These observations suggest that the high mtDNA copy number in the mature oocyte is a genetic device designed to distribute mitochondria and mtDNAs to the cells of the early postimplantation embryo before mitochondrial biogenesis and mtDNA replication resumes, whereas down-regulation of mtDNA copy number is important for normal sperm function.


Asunto(s)
ADN Mitocondrial/metabolismo , Desarrollo Embrionario , Fertilidad , Dosificación de Gen , Oocitos/metabolismo , Animales , Femenino , Fertilización , Masculino , Ratones , Ratones Noqueados , Recuento de Espermatozoides , Motilidad Espermática
20.
Fertil Steril ; 94(6): 2026-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20171615

RESUMEN

OBJECTIVE: To evaluate the association between endometrial injury, implantation and pregnancy rate. DESIGN: We performed a literature search using the keywords endometrial injury, local endometrial injury, endometrial biopsy, endometrial receptivity, implantation, in vitro fertilization, and implantation failure and conducted the search in Medline, EMBASE, and Cochrane Database of systematic reviews. SETTING: None. PATIENT(S): None. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): None. RESULT(S): Clinical and basic science data regarding the association between endometrial injury and improved implantation rate are limited. However, current evidence suggests that endometrial injury before IVF among women with previous repeated IVF failure is associated with increased rates of implantation, clinical pregnancy, and live birth. CONCLUSION(S): Endometrial injury may have a beneficial role in implantation and improve the pregnancy rate. However, there are still many unanswered question including patients selection, timing, technique and number of endometrial biopsies needed.


Asunto(s)
Implantación del Embrión/fisiología , Endometrio/lesiones , Endometrio/cirugía , Infertilidad Femenina/cirugía , Aborto Espontáneo/epidemiología , Aborto Espontáneo/etiología , Animales , Ensayos Clínicos como Asunto/estadística & datos numéricos , Endometrio/patología , Femenino , Humanos , Infertilidad Femenina/epidemiología , Infertilidad Femenina/patología , Embarazo , Resultado del Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA