Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Dev Biol ; 58(2-4): 79-86, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25023673

RESUMO

This interview chronicles the story of Sudhansu K. Dey in his journey from Calcutta, India to Kansas City, Kansas, establishing a research enterprise in the field of female reproduction. His research of over four decades has focused specifically on implantation biology using various model systems and reveling the impact of implantation on female reproductive medicine. This interview also reveals qualities of SK's character - his resolution, mentoring spirit, and humble nature - that contributed to his successes. SK is not shy to approach individuals for expertise or help, and in the same spirit, he is ready to offer his help to others irrespective of their positions or stature. He constantly attributes his success to the hard work of his laboratory members, the intellectual stimulation from his collaborators, and the support from his family. His ability to overcome challenges throughout his career is a reminder to students and junior investigators in the scientific community that each individual is endowed with talents and can accomplish their dreams if they pursue them. This interview tells the story of how he progressed from an inquisitive child to becoming a true servant to the cause of science and humankind.


Assuntos
Pesquisa Biomédica , Implantação do Embrião/fisiologia , Embrião de Mamíferos/fisiologia , Reprodução/fisiologia , Feminino , Humanos
2.
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
3.
Proc Natl Acad Sci U S A ; 107(35): 15577-82, 2010 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-20713718

RESUMO

Immunophilin FK506-binding protein 52 (FKBP52) is a cochaperone that binds to the progesterone receptor (PR) to optimize progesterone (P(4))-PR signaling. We recently showed that Fkbp52-deficient (Fkbp52(-/-)) mice have reduced uterine PR responsiveness and implantation failure which is rescued by excess P(4) supplementation in a genetic background-dependent manner. This finding led us to hypothesize that FKBP52 has functions in addition to optimizing PR activity. Using proteomics analysis, we found that uterine levels of peroxiredoxin-6 (PRDX6), a unique antioxidant, are significantly lower in Fkbp52(-/-) mice than in WT and PR-null (Pgr(-/-)) mice. We also found that Fkbp52(-/-) mice with reduced uterine PRDX6 levels are susceptible to paraquat-induced oxidative stress (OS), leading to implantation failure even with P(4) supplementation. The same dose of paraquat did not interfere with implantation in WT mice. Moreover, treatment with antioxidants alpha-tocopherol and N-acetylcysteine (NAC) attenuated paraquat-induced implantation failure in P(4)-treated Fkbp52(-/-) mice. Functional analyses using mouse embryonic fibroblasts show that Fkbp52 deficiency associated with reduced PRDX6 levels promotes H(2)O(2)-induced cell death, which is reversed by the addition of NAC or by forced expression of PRDX6, suggesting that Fkbp52 deficiency diminishes the threshold against OS by reducing PRDX6 levels. These findings provide evidence that heightened uterine OS in Fkbp52(-/-) females with reduced PRDX6 levels induces implantation failure even in the presence of excess P(4). This study shows that FKBP52-PRDX6 signaling protects pregnancy from overt OS.


Assuntos
Estresse Oxidativo , Peroxirredoxina VI/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Ligação a Tacrolimo/metabolismo , Útero/metabolismo , Animais , Northern Blotting , Western Blotting , Implantação do Embrião/efeitos dos fármacos , Endométrio/citologia , Endométrio/metabolismo , Feminino , Perfilação da Expressão Gênica , Herbicidas/farmacologia , Humanos , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovariectomia , Paraquat/farmacologia , Peroxirredoxina VI/genética , Gravidez , Progesterona/farmacologia , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Fatores de Tempo , Útero/efeitos dos fármacos
4.
J Clin Invest ; 120(3): 803-15, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20124728

RESUMO

Many signaling pathways that contribute to tumorigenesis are also functional in pregnancy, although they are dysregulated in the former and tightly regulated in the latter. Transformation-related protein 53 (Trp53), which encodes p53, is a tumor suppressor gene whose mutation is strongly associated with cancer. However, its role in normal physiological processes, including female reproduction, is poorly understood. Mice that have a constitutive deletion of Trp53 exhibit widespread development of carcinogenesis at early reproductive ages, compromised spermatogenesis, and fetal exencephaly, rendering them less amenable to studying the role of p53 in reproduction. To overcome this obstacle, we generated mice that harbor a conditional deletion of uterine Trp53 and examined pregnancy outcome in females with this genotype. These mice had normal ovulation, fertilization, and implantation; however, postimplantation uterine decidual cells showed terminal differentiation and senescence-associated growth restriction with increased levels of phosphorylated Akt and p21, factors that are both known to participate in these processes in other systems. Strikingly, uterine deletion of Trp53 increased the incidence of preterm birth, a condition that was corrected by oral administration of the selective COX2 inhibitor celecoxib. We further generated evidence to suggest that deletion of uterine Trp53 induces preterm birth through a COX2/PGF synthase/PGF(2alpha) pathway. Taken together, our observations underscore what we believe to be a new critical role of uterine p53 in parturition.


Assuntos
Decídua/metabolismo , Nascimento Prematuro/metabolismo , Proteína Supressora de Tumor p53 , Animais , Celecoxib , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Decídua/patologia , Dinoprosta/genética , Dinoprosta/metabolismo , Implantação do Embrião/efeitos dos fármacos , Implantação do Embrião/genética , Feminino , Fertilização/efeitos dos fármacos , Fertilização/genética , Hidroxiprostaglandina Desidrogenases/genética , Hidroxiprostaglandina Desidrogenases/metabolismo , Camundongos , Camundongos Transgênicos , Ovulação/efeitos dos fármacos , Ovulação/genética , Ovulação/metabolismo , Gravidez , Nascimento Prematuro/genética , Nascimento Prematuro/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirazóis/farmacologia , Sulfonamidas/farmacologia
5.
J Lipid Res ; 50(11): 2290-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19429885

RESUMO

Molecular events involved in successful embryo implantation are not well understood. In this study, we used MALDI imaging mass spectrometry (IMS) technologies to characterize the spatial and temporal distribution of phospholipid species associated with mouse embryo implantation. Molecular images showing phospholipid distribution within implantation sites changed markedly between distinct cellular areas during days 4-8 of pregnancy. For example, by day 8, linoleate- and docosahexaenoate-containing phospholipids localized to regions destined to undergo cell death, whereas oleate-containing phospholipids localized to angiogenic regions. Arachidonate-containing phospholipids showed different segregation patterns depending on the lipid class, revealing a strong correlation of phosphatidylethanolamines and phosphatidylinositols with cytosolic phospholipase A(2alpha) and cyclooxygenase-2 during embryo implantation. LC-ESI-MS/MS was used to validate MALDI IMS phospholipid distribution patterns. Overall, molecular images revealed the dynamic complexity of lipid distributions in early pregnancy, signifying the importance of complex interplay of lipid molecules in uterine biology and implantation.


Assuntos
Implantação do Embrião , Espectrometria de Massas/métodos , Fosfolipídeos/análise , Animais , Ciclo-Oxigenase 2/metabolismo , Citosol/enzimologia , Feminino , Fosfolipases A2 do Grupo IV/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Fosfatidilcolinas/análise , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/análise , Fosfatidiletanolaminas/metabolismo , Fosfatidilinositóis/análise , Fosfatidilinositóis/metabolismo , Fosfolipídeos/metabolismo , Gravidez , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Esfingomielinas/análise , Esfingomielinas/metabolismo , Fatores de Tempo , Útero/metabolismo
6.
Am J Pathol ; 173(6): 1747-57, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18988805

RESUMO

Endometriosis is a common gynecological disease that affects approximately 10% of women of childbearing age. It is characterized by endometrial growth outside the uterus and often results in inflamed lesions, pain, and reduced fertility. Although heightened estrogenic activity and/or reduced progesterone responsiveness are considered to be involved in the etiology of endometriosis, neither the extent of their participation nor the underlying mechanisms are clearly understood. Heterogeneous uterine cell types differentially respond to estrogen and progesterone (P(4)). P(4), primarily acting via its nuclear receptor (PR), activates gene transcription and impacts many reproductive processes. Deletion of Fkbp52, an immunophilin cochaperone for PR, results in uterine-specific P(4) resistance in mice, creating an opportunity to study the unique aspects of P(4) signaling in endometriosis. Here we explored the roles of FKBP52 in this disease using Fkbp52(-/-) mice. We found that the loss of FKBP52 encourages the growth of endometriotic lesions with increased inflammation, cell proliferation, and angiogenesis. We also found remarkable down-regulation of FKBP52 in cases of human endometriosis. Our results provide the first evidence corroborated by genetic studies in mice for a potential role of an immunophilin cochaperone in the etiology of human endometriosis. This investigation is highly relevant for clinical application, particularly because P(4) resistance is favorably indicated in endometriosis and other gynecological diseases.


Assuntos
Endometriose/metabolismo , Proteínas de Ligação a Tacrolimo/deficiência , Animais , Proliferação de Células , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Endometriose/patologia , Endometriose/fisiopatologia , Endométrio/metabolismo , Endométrio/patologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neovascularização Patológica , Progesterona/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Development ; 135(22): 3697-706, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18927152

RESUMO

Meiosis is coupled to gamete development and must be well regulated to prevent aneuploidy. During meiotic maturation, Drosophila oocytes progress from prophase I to metaphase I. The molecular factors controlling meiotic maturation timing, however, are poorly understood. We show that Drosophila alpha-endosulfine (endos) plays a key role in this process. endos mutant oocytes have a prolonged prophase I and fail to progress to metaphase I. This phenotype is similar to that of mutants of cdc2 (synonymous with cdk1) and of twine, the meiotic homolog of cdc25, which is required for Cdk1 activation. We found that Twine and Polo kinase levels are reduced in endos mutants, and identified Early girl (Elgi), a predicted E3 ubiquitin ligase, as a strong Endos-binding protein. In elgi mutant oocytes, the transition into metaphase I occurs prematurely, but Polo and Twine levels are unaffected. These results suggest that Endos controls meiotic maturation by regulating Twine and Polo levels, and, independently, by antagonizing Elgi. Finally, germline-specific expression of the human alpha-endosulfine ENSA rescues the endos mutant meiotic defects and infertility, and alpha-endosulfine is expressed in mouse oocytes, suggesting potential conservation of its meiotic function.


Assuntos
Diferenciação Celular , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Meiose , Oócitos/citologia , Oócitos/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Evolução Biológica , Proteína Quinase CDC2/metabolismo , Sequência Conservada , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Dados de Sequência Molecular , Mutação/genética , Membrana Nuclear , Peptídeos/química , Peptídeos/genética , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Alinhamento de Sequência , Ubiquitina-Proteína Ligases/metabolismo
8.
Cancer Res ; 68(14): 5619-27, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18632614

RESUMO

Etiology of endometrial cancer (EMC) is not fully understood. Animal models with rapidly and spontaneously developing EMC will help explore mechanisms of cancer initiation and progression. Pten(+/-) mice are currently being used as a model to study EMC. These females develop atypical endometrial hyperplasia of which approximately 20% progresses to EMC. In addition, tumors develop in other organs, complicating the use of this model to specifically study EMC. Here, we show that conditional deletion of endometrial Pten results in EMC in all female mice as early as age 1 month with myometrial invasion occurring by 3 months. In contrast, conditional deletion of endometrial p53 had no phenotype within this time frame. Whereas mice with endometrial Pten deletion had a life span of approximately 5 months, mice with combined deletion of endometrial Pten and p53 had a shorter life span with an exacerbated disease state. Such rapid development of EMC from homozygous loss of endometrial Pten suggests that this organ is very sensitive to this tumor suppressor gene for tumor development. All lesions at early stages exhibited elevated Cox-2 and phospho-Akt levels, hallmarks of solid tumors. More interestingly, levels of two microRNAs miR-199a(*) and miR-101a that posttranscriptionally inhibit Cox-2 expression were down-regulated in tumors in parallel with Cox-2 up-regulation. This mouse model in which the loxP-Cre system has been used to delete endometrial Pten and/or p53 allows us to study in detail the initiation and progression of EMC. These mouse models have the added advantage because they mimic several features of human EMC.


Assuntos
Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Regulação Neoplásica da Expressão Gênica , PTEN Fosfo-Hidrolase/fisiologia , Útero/metabolismo , Animais , Ciclo-Oxigenase 2/genética , Feminino , Genes p53 , Homozigoto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/genética , PTEN Fosfo-Hidrolase/genética , Fenótipo , Processamento Pós-Transcricional do RNA
9.
Endocrinology ; 149(7): 3274-8, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18403475

RESUMO

A reciprocal interaction between the implantation-competent blastocyst and receptive uterus is an absolute requirement for implantation, a process crucial for pregnancy success. A comprehensive understanding of this interaction has yet to be realized. One major difficulty in clearly defining this discourse is the complexity of the implantation process involving heterogeneous cell types of both the uterus and blastocyst, each endowed with unique molecular signatures that show dynamic changes during the course of pregnancy. Whereas gene expression studies by in situ hybridization or immunohistochemistry have shown differential expression patterns of specific genes during implantation, there is no report how numerous signaling proteins are spatially displayed at specific times and stages of implantation in the context of blastocyst-uterine juxtaposition. Using in situ imaging (matrix assisted laser desorption/ionization) mass spectrometry directly on uterine sections, here we provide molecular composition, relative abundance, and spatial distribution of a large number of proteins during the periimplantation period. This approach has allowed us for the first time to generate in situ proteome profiles of implantation and interimplantation sites in mice in a region- and stage-specific manner with the progression of implantation. This application is reliable because patterns of expression of several proteins displayed by in situ imaging mass spectrometry correlate well with in situ hybridization results. More interestingly, the use of this approach has provided new insights regarding uterine biology of cytosolic phospholipase A(2alpha) null females that show implantation defects.


Assuntos
Implantação do Embrião/fisiologia , Espectrometria de Massas/métodos , Proteínas/metabolismo , Animais , Blastocisto/metabolismo , Implantação do Embrião/genética , Feminino , Perfilação da Expressão Gênica , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Gravidez , Proteínas/genética , Proteômica/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Útero/metabolismo
10.
Development ; 135(4): 717-27, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18199579

RESUMO

The activation of the blastocyst, a process by which it gains competency to attach with the receptive uterus, is a prerequisite for successful implantation. However, the molecular basis of blastocyst activation remains largely unexplored. Combining molecular, pharmacological and physiological approaches, we show here that silencing of Wnt-beta-catenin signaling in mice does not adversely affect the development of preimplantation embryos to blastocysts and uterine preparation for receptivity, but, remarkably, blocks blastocyst competency to implantation. Using the physiologically relevant delayed implantation model and trophoblast stem cells in culture, we further demonstrate that a coordinated activation of canonical Wnt-beta-catenin signaling with attenuation of the non-canonical Wnt-RhoA signaling pathway ensures blastocyst competency to implantation. These findings constitute novel evidence that Wnt signaling is at least one pathway that determines blastocyst competency for implantation.


Assuntos
Blastocisto/citologia , Blastocisto/metabolismo , Núcleo Celular/metabolismo , Implantação do Embrião , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Células Cultivadas , Transferência Embrionária , Feminino , Inativação Gênica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , PPAR delta/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Trofoblastos/citologia , Trofoblastos/metabolismo , Proteína Wnt3
11.
Proc Natl Acad Sci U S A ; 104(46): 18315-20, 2007 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-17986609

RESUMO

An intimate discourse between the blastocyst and uterus is essential for successful implantation. However, the molecular basis of this interaction is not clearly understood. Exploiting genomic Hbegf mutant mice, we show here that maternal deficiency of heparin-binding EGF-like growth factor (HB-EGF) defers on-time implantation, leading to compromised pregnancy outcome. We also demonstrate that amphiregulin, but not epiregulin, partially compensates for the loss of HB-EGF during implantation. In search of the mechanism of this compensation, we found that reduced preimplantation estrogen secretion from ovarian HB-EGF deficiency is a cause of sustained expression of uterine amphiregulin before the initiation of implantation. To explore the significance specifically of uterine HB-EGF in implantation, we examined this event in mice with conditional deletion of uterine HB-EGF and found that this specific loss of HB-EGF in the uterus still defers on-time implantation without altering preimplantation ovarian estrogen secretion. The observation of normal induction of uterine amphiregulin surrounding the blastocyst at the time of attachment in these conditional mutant mice suggests a compensatory role of amphiregulin for uterine loss of HB-EGF, preventing complete failure of pregnancy. Our study provides genetic evidence that HB-EGF is critical for normal implantation. This finding has high clinical relevance, because HB-EGF signaling is known to be important for human implantation.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Animais , Feminino , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Tamanho da Ninhada de Vivíparos , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
12.
J Biol Chem ; 282(52): 37770-82, 2007 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-17965409

RESUMO

Successful pregnancy depends on well coordinated developmental events involving both maternal and embryonic components. Although a host of signaling pathways participate in implantation, decidualization, and placentation, whether there is a common molecular link that coordinates these processes remains unknown. By exploiting genetic, molecular, pharmacological, and physiological approaches, we show here that the nuclear transcription factor peroxisome proliferator-activated receptor (PPAR) delta plays a central role at various stages of pregnancy, whereas maternal PPARdelta is critical to implantation and decidualization, and embryonic PPARdelta is vital for placentation. Using trophoblast stem cells, we further elucidate that a reciprocal relationship between PPARdelta-AKT and leukemia inhibitory factor-STAT3 signaling pathways serves as a cell lineage sensor to direct trophoblast cell fates during placentation. This novel finding of stage-specific integration of maternal and embryonic PPARdelta signaling provides evidence that PPARdelta is a molecular link that coordinates implantation, decidualization, and placentation crucial to pregnancy success. This study is clinically relevant because deferral of on time implantation leads to spontaneous pregnancy loss, and defective trophoblast invasion is one cause of preeclampsia in humans.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , PPAR delta/metabolismo , Prenhez/fisiologia , Animais , Decídua/metabolismo , Implantação do Embrião , Feminino , Fertilização , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovulação , Placenta/metabolismo , Gravidez , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais , Células-Tronco/citologia , Fatores de Tempo , Trofoblastos/metabolismo
13.
Proc Natl Acad Sci U S A ; 104(38): 15144-9, 2007 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-17848513

RESUMO

The implantation process is complex, requiring reciprocal interactions between implantation-competent blastocysts and the receptive uterus. Because microRNAs (miRNAs) have major roles in regulating gene expression, we speculated that they participate in directing the highly regulated spatiotemporally expressed genetic network during implantation. Here, we show that two miRNAs, mmu-miR-101a and mmu-miR-199a*, are spatiotemporally expressed in the mouse uterus during implantation coincident with expression of cyclooxygenase-2, a gene critical for implantation. More interestingly, our in vitro gain- and loss-of-function experiments show that cyclooxygenase-2 expression is posttranscriptionally regulated by these two miRNAs. We report on miRNA-mediated regulation of uterine gene expression in the context of implantation. We believe that many other critical genes related to this process are also regulated by miRNAs. Thus, elucidating the physiological roles of uterine miRNAs will help us better understand the genetic control of implantation, the gateway to a successful pregnancy.


Assuntos
Ciclo-Oxigenase 2/genética , Implantação do Embrião/genética , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/metabolismo , Útero/metabolismo , Animais , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos , Gravidez
14.
J Clin Invest ; 117(7): 1824-34, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17571166

RESUMO

Immunophilin FKBP52 serves as a cochaperone to govern normal progesterone (P(4)) receptor (PR) function. Using Fkbp52(-/-) mice, we show intriguing aspects of uterine P(4)/PR signaling during pregnancy. Implantation failure is the major phenotype found in these null females, which is conserved on both C57BL6/129 and CD1 backgrounds. However, P(4) supplementation rescued implantation and subsequent decidualization in CD1, but not C57BL6/129, null females. Surprisingly, experimentally induced decidualization in the absence of blastocysts failed in Fkbp52(-/-) mice on either background even with P(4) supplementation, suggesting that embryonic signals complement uterine signaling for this event. Another interesting finding was that while P(4) at higher than normal pregnancy levels conferred PR signaling sufficient for implantation in CD1 null females, these levels were inefficient in maintaining pregnancy to full term. However, elevating P(4) levels further restored PR signaling to a level optimal for successful term pregnancy with normal litter size. Collectively, the results show that the indispensability of FKBP52 in uterine P(4)/PR signaling is a function of genetic disparity and is pregnancy stage specific. Since there is evidence for a correlation between P(4) supplementation and reduced risks of P(4)-resistant recurrent miscarriages and remission of endometriosis, these findings have clinical implications for genetically diverse populations of women.


Assuntos
Resistência a Medicamentos , Progesterona/farmacologia , Proteínas de Ligação a Tacrolimo/deficiência , Proteínas de Ligação a Tacrolimo/metabolismo , Útero/efeitos dos fármacos , Útero/metabolismo , Animais , Blastocisto/efeitos dos fármacos , Blastocisto/metabolismo , Transferência Embrionária , Feminino , Camundongos , Camundongos Knockout , Gravidez , Receptores de Progesterona/metabolismo , Transdução de Sinais , Proteínas de Ligação a Tacrolimo/genética
15.
Biol Reprod ; 77(3): 425-32, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17538075

RESUMO

Reduced litter sizes in mice missing pentraxin 3 (Ptx3) have been attributed to fertilization failure. However, our global gene expression studies showed high uterine Ptx3 expression at the implantation site in mice, suggesting its role in blastocyst implantation. We initiated molecular and genetic studies in mice to explore the importance of uterine Ptx3 in this process. We found that Ptx3 is expressed in a unique and transient fashion at implantation sites. With the initiation of implantation on midnight of Day 4 of pregnancy, Ptx3 is expressed exclusively in stromal cells at the site of blastocysts. On Day 5, its expression is more intense in decidualizing stromal cells, but it disappears on Day 6. The expression again becomes evident in the deciduum on Day 7, followed by a more robust expression on Day 8, particularly at the antimesometrial pole. From Day 9, with the initiation of placentation, Ptx3 expression becomes undetectable. These results suggest a role for PTX3 in implantation and decidualization. Indeed, deletion of Ptx3 results in both compromised implantation and decidualization. Interleukin 1B (IL1B), a known inducer of Ptx3, is also transiently expressed in stromal cells at the implantation site, suggesting that IL1B is an inducer of uterine Ptx3 expression. In fact, uterine Ptx3 expression follows that of Il1b induced by lipopolysaccharide treatment on Day 7 of pregnancy. Collectively, these findings provide evidence for an important role for PTX3 in implantation and decidualization. This study has clinical implications, since PTX3 is expressed in the receptive endometrium, and trophoblast cells influence decidual Ptx3 expression in humans.


Assuntos
Proteína C-Reativa/deficiência , Implantação do Embrião/fisiologia , Proteínas do Tecido Nervoso/deficiência , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Northern Blotting , Proteína C-Reativa/biossíntese , Proteína C-Reativa/genética , Decídua/citologia , Decídua/metabolismo , Feminino , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Útero/metabolismo , Útero/fisiologia
16.
Cancer Res ; 67(11): 5285-92, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17545608

RESUMO

The underlying causes of epithelial ovarian cancer (EOC) are unclear, and treatment options for patients with advanced disease are limited. There is evidence that the use of nonsteroidal anti-inflammatory drugs is associated with decreased risk of developing EOC. Nonsteroidal anti-inflammatory drugs inhibit cyclooxygenase (COX)-1 and COX-2, which catalyze prostaglandin biosynthesis. We previously showed that mouse and human EOCs have increased levels of COX-1, but not COX-2, and a COX-1-selective inhibitor, SC-560, attenuates prostaglandin production and tumor growth. However, the downstream targets of COX-1 signaling in EOC are not yet known. To address this question, we evaluated peroxisome proliferator-activated receptor delta (PPARdelta) expression and function in EOC. We found that EOC cells express high levels of PPARdelta, and neutralizing PPARdelta function reduces tumor growth in vivo. More interestingly, aspirin, a nonsteroidal anti-inflammatory drug that preferentially inhibits COX-1, compromises PPARdelta function and cell growth by inhibiting extracellular signal-regulated kinases 1/2, members of the mitogen-activated protein kinase family. Our study, for the first time, shows that whereas PPARdelta can be a target of COX-1, extracellular signal-regulated kinase is a potential target of PPARdelta. The ability of aspirin to inhibit EOC growth in vivo is an exciting finding because of its low cost, lack of cardiovascular side effects, and availability.


Assuntos
Ciclo-Oxigenase 1/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neoplasias Ovarianas/metabolismo , PPAR delta/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Inibidores de Ciclo-Oxigenase/farmacologia , Ativação Enzimática , Células Epiteliais/patologia , Epoprostenol/metabolismo , Feminino , Humanos , Masculino , Camundongos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , PPAR delta/antagonistas & inibidores , PPAR delta/biossíntese , Pirazóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Reproduction ; 133(2): 395-403, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17307907

RESUMO

FKBP52 is a member of the FK506-binding family of immunophilins and serves as a co-chaperone for steroid hormone nuclear receptors to govern appropriate hormone action in target tissues. Male mice missing Fkbp52 are infertile, and this infertility has been ascribed to compromised sensitivity of the anterior prostate, external genitalia, and other accessory sex organs to androgen. Here, we show additional defects contributing to infertility. We found that epididymal Fkbp52(-/-) sperm are sparse often with aberrant morphology, and they have reduced fertilizing capacity. This phenotype, initially observed in null males on a C57BL/6/129 background, is also maintained on a CD1 background. Expression studies show that while FKBP52 and androgen receptor are co-expressed in similar cell types in the epididymis, FKBP52 is also present in epididymal sperm flagella. Collectively, our results suggest that reduced number and abnormal morphology contribute to compromised fertilizing capacity of Fkbp52(-/-) sperm. This study is clinically relevant because unraveling the role of immunophilin signaling in male fertility will help identify new targets for male contraceptives and/or alleviate male infertility.


Assuntos
Reação Acrossômica/fisiologia , Chaperonas Moleculares , Espermatozoides/metabolismo , Proteínas de Ligação a Tacrolimo/fisiologia , Animais , Western Blotting , Epididimo/química , Epididimo/metabolismo , Feminino , Fertilização in vitro , Técnica Indireta de Fluorescência para Anticorpo , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Androgênicos/análise , Receptores Androgênicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cauda do Espermatozoide/química , Cauda do Espermatozoide/metabolismo , Espermatozoides/citologia , Proteínas de Ligação a Tacrolimo/análise , Proteínas de Ligação a Tacrolimo/genética
18.
Reprod Biomed Online ; 14 Spec No 1: 39-48, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-20483398

RESUMO

Embryo implantation is absolutely dependent on the preparation of the uterus to the receptive stage and attainment by the blastocyst of implantation competency. Co-ordinated effects of progesterone and oestrogen are essential for these processes and determine the window of implantation. In rodents, a generalized stromal edema occurs before blastocyst attachment followed by uterine luminal closure. This leads to apposition of the blastocyst trophectoderm against the luminal epithelium and ultimately attachment. Progesterone is essential for luminal closure, which must occur for successful implantation. More importantly, progesterone is critical for almost every stage of pregnancy, including ovulation, fertilization, implantation, decidualization and pregnancy maintenance. Progesterone exerts its effects on target tissues primarily via nuclear progesterone receptor (PR) whose optimal activity is potentiated by an immunophilin co-chaperone, FK-506 binding protein 4 (FKBP52). While mice lacking PR are infertile due to complete failure of ovulation, fertilization, and implantation, female mice with targeted deletion of the Fkbp52 gene are infertile specifically because of implantation failure resulting from compromised uterine receptivity. This review highlights the evolution of knowledge about progesterone signalling during early pregnancy. Future studies are likely to provide a better understanding of FKBP52-PR signalling in promoting uterine receptivity for implantation and may reveal new targets for improving infertility.

19.
Reprod Biomed Online ; 13(5): 651-60, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17169175

RESUMO

Embryo implantation is absolutely dependent on the preparation of the uterus to the receptive stage and attainment by the blastocyst of implantation competency. Co-ordinated effects of progesterone and oestrogen are essential for these processes and determine the window of implantation. In rodents, a generalized stromal edema occurs before blastocyst attachment followed by uterine luminal closure. This leads to apposition of the blastocyst trophectoderm against the luminal epithelium and ultimately attachment. Progesterone is essential for luminal closure, which must occur for successful implantation. More importantly, progesterone is critical for almost every stage of pregnancy, including ovulation, fertilization, implantation, decidualization and pregnancy maintenance. Progesterone exerts its effects on target tissues primarily via nuclear progesterone receptor (PR) whose optimal activity is potentiated by an immunophilin co-chaperone, FK-506 binding protein 4 (FKBP52). While mice lacking PR are infertile due to complete failure of ovulation, fertilization, and implantation, female mice with targeted deletion of the Fkbp52 gene are infertile specifically because of implantation failure resulting from compromised uterine receptivity. This review highlights the evolution of knowledge about progesterone signalling during early pregnancy. Future studies are likely to provide a better understanding of FKBP52-PR signalling in promoting uterine receptivity for implantation and may reveal new targets for improving infertility.


Assuntos
Manutenção da Gravidez/fisiologia , Progesterona/fisiologia , Receptores de Progesterona/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Corpo Lúteo/fisiologia , Implantação do Embrião/fisiologia , Feminino , Camundongos , Gravidez , Transdução de Sinais
20.
Biochem J ; 399(1): 91-9, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16787386

RESUMO

COX [cyclo-oxygenase; PG (prostaglandin) G/H synthase] oxygenates AA (arachidonic acid) and 2-AG (2-arachidonylglycerol) to endoperoxides that are converted into PGs and PG-Gs (glycerylprostaglandins) respectively. In vitro, 2-AG is a selective substrate for COX-2, but in zymosan-stimulated peritoneal macrophages, PG-G synthesis is not sensitive to selective COX-2 inhibition. This suggests that COX-1 oxygenates 2-AG, so studies were carried out to identify enzymes involved in zymosan-dependent PG-G and PG synthesis. When macrophages from COX-1-/- or COX-2-/- mice were treated with zymosan, 20-25% and 10-15% of the PG and PG-G synthesis observed in wild-type cells respectively was COX-2 dependent. When exogenous AA and 2-AG were supplied to COX-2-/- macrophages, PG and PG-G synthesis was reduced as compared with wild-type cells. In contrast, when exogenous substrates were provided to COX-1-/- macrophages, PG-G but not PG synthesis was reduced. Product synthesis also was evaluated in macrophages from cPLA(2alpha) (cytosolic phospholipase A2alpha)-/- mice, in which zymosan-induced PG synthesis was markedly reduced, and PG-G synthesis was increased approx. 2-fold. These studies confirm that peritoneal macrophages synthesize PG-Gs in response to zymosan, but that this process is primarily COX-1-dependent, as is the synthesis of PGs. They also indicate that the 2-AG and AA used for PG-G and PG synthesis respectively are derived from independent pathways.


Assuntos
Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Macrófagos Peritoneais/enzimologia , Prostaglandinas/biossíntese , Zimosan/farmacologia , Animais , Células Cultivadas , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/genética , Regulação Enzimológica da Expressão Gênica , Fosfolipases A2 do Grupo IV , Macrófagos Peritoneais/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Fosfolipases A/genética , Fosfolipases A/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...