Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Physiol Rev ; 103(3): 1965-2038, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-36796099

RESUMO

Pregnancy is established during the periconceptional period as a continuum beginning with blastocyst attachment to the endometrial epithelial surface followed by embryo invasion and placenta formation. This period sets the foundation for the child and mother's health during pregnancy. Emerging evidence indicates that prevention of downstream pathologies in both the embryo/newborn and pregnant mother may be possible at this stage. In this review, we discuss current advances in the periconceptional space, including the preimplantation human embryo and maternal endometrium. We also discuss the role of the maternal decidua, the periconceptional maternal-embryonic interface, the dialogue between these elements, and the importance of the endometrial microbiome in the implantation process and pregnancy. Finally, we discuss the myometrium in the periconceptional space and review its role in determining pregnancy health.


Assuntos
Implantação do Embrião , Endométrio , Gravidez , Feminino , Criança , Recém-Nascido , Humanos , Blastocisto , Placenta
2.
Am J Obstet Gynecol ; 226(2S): S886-S894, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-33007270

RESUMO

Preeclampsia is a major obstetrical complication with short- and long-term life-threatening consequences for both mother and child. Shallow cytotrophoblast invasion through the uterine decidua into the spiral arteries is implicated in the pathogenesis of preeclampsia, although the cause of deficient arterial invasion remains unknown. Research that is focused on the "soil"-the maternal decidua-highlights the importance of this poorly understood but influential uterine layer. Decidualization of endometrial cells regulates embryo invasion, which is essential for spiral artery remodeling and establishing the maternal-fetal interface. Exploration of the association between impaired decidualization and preeclampsia revealed suboptimal endometrial maturation and uterine natural killer cells present in the decidua before preeclampsia development. Furthermore, decidualization defects in the endometrium of women with severe preeclampsia, characterized by impaired cytotrophoblast invasion, were detected at the time of delivery and persisted 5 years after the affected pregnancy. Recently, a maternal deficiency of annexin A2 expression was found to influence aberrant decidualization and shallow cytotrophoblast invasion, suggesting that decidualization resistance, which is a defective endometrial cell differentiation during the menstrual cycle, could underlie shallow trophoblast invasion and the poor establishment of the maternal-fetal interface. Based on these findings, the transcriptional signature in the endometrium that promotes decidualization deficiency could be detected before (or after) conception. This would serve to identify women at risk of developing severe preeclampsia and aid the development of therapies focused on improving decidualization, perhaps also preventing severe preeclampsia. Here, we discuss decidualization deficiency as a contributor to the pathogenesis of pregnancy disorders with particular attention to severe preeclampsia. We also review current diagnostic strategies and discuss future directions in diagnostic methods based on decidualization.


Assuntos
Decídua/fisiopatologia , Pré-Eclâmpsia/fisiopatologia , Anexina A2/genética , Anexina A2/metabolismo , Decídua/metabolismo , Diagnóstico Precoce , Endométrio/patologia , Feminino , Humanos , Placentação/fisiologia , Pré-Eclâmpsia/diagnóstico , Gravidez , Trofoblastos/fisiologia
3.
Elife ; 102021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34709177

RESUMO

Background: Decidualization of the uterine mucosa drives the maternal adaptation to invasion by the placenta. Appropriate depth of placental invasion is needed to support a healthy pregnancy; shallow invasion is associated with the development of severe preeclampsia (sPE). Maternal contribution to sPE through failed decidualization is an important determinant of placental phenotype. However, the molecular mechanism underlying the in vivo defect linking decidualization to sPE is unknown. Methods: Global RNA sequencing was applied to obtain the transcriptomic profile of endometrial biopsies collected from nonpregnant women who suffer sPE in a previous pregnancy and women who did not develop this condition. Samples were randomized in two cohorts, the training and the test set, to identify the fingerprinting encoding defective decidualization in sPE and its subsequent validation. Gene Ontology enrichment and an interaction network were performed to deepen in pathways impaired by genetic dysregulation in sPE. Finally, the main modulators of decidualization, estrogen receptor 1 (ESR1) and progesterone receptor B (PGR-B), were assessed at the level of gene expression and protein abundance. Results: Here, we discover the footprint encoding this decidualization defect comprising 120 genes-using global gene expression profiling in decidua from women who developed sPE in a previous pregnancy. This signature allowed us to effectively segregate samples into sPE and control groups. ESR1 and PGR were highly interconnected with the dynamic network of the defective decidualization fingerprint. ESR1 and PGR-B gene expression and protein abundance were remarkably disrupted in sPE. Conclusions: Thus, the transcriptomic signature of impaired decidualization implicates dysregulated hormonal signaling in the decidual endometria in women who developed sPE. These findings reveal a potential footprint that could be leveraged for a preconception or early prenatal screening of sPE risk, thus improving prevention and early treatments. Funding: This work has been supported by the grant PI19/01659 (MCIU/AEI/FEDER, UE) from the Spanish Carlos III Institute awarded to TGG. NCM was supported by the PhD program FDGENT/2019/008 from the Spanish Generalitat Valenciana. IMB was supported by the PhD program PRE2019-090770 and funding was provided by the grant RTI2018-094946-B-100 (MCIU/AEI/FEDER, UE) from the Spanish Ministry of Science and Innovation with CS as principal investigator. This research was funded partially by Igenomix S.L.


Assuntos
Decídua/patologia , Receptor alfa de Estrogênio/genética , Pré-Eclâmpsia/genética , Receptores de Progesterona/genética , Transdução de Sinais , Adulto , Decídua/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Pré-Eclâmpsia/metabolismo , Gravidez , Receptores de Progesterona/metabolismo , Adulto Jovem
4.
Am J Obstet Gynecol ; 222(4): 376.e1-376.e17, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31738896

RESUMO

BACKGROUND: Decidualization defects in the endometrium have been demonstrated at the time of delivery in women with severe preeclampsia and to linger for years, which suggests a maternal contribution to the pathogenesis of this condition. Global transcriptional profiling reveals alterations in gene expression, which includes down-regulation of Annexin A2 in severe preeclampsia patients with decidualization resistance. OBJECTIVE: We investigated the functional role of Annexin A2 deficiency during endometrial decidualization and its potential contribution to shallow trophoblast invasion during implantation and subsequent placentation using in vitro and in vivo modeling. STUDY DESIGN: Annexin A2 gene and protein levels were assessed during in vitro decidualization of human endometrial stromal cells isolated from biopsy specimens that were collected from women with previous severe preeclampsia (n=5) or normal obstetric outcomes (n=5). Next, Annexin A2 was inhibited with small interference RNA in control human endometrial stromal cells that were isolated from endometrial biopsy specimens (n=15) as an in vitro model to analyze decidualization defects at the morphologic level and the secretion of prolactin and insulin-like growth binding protein-1. Annexin A2-inhibited cells were used to evaluate motility and promotion of embryo invasion. Decidualization and placentation defects of Annexin A2 deficiency were confirmed with the use of an Annexin A2-null mouse model. RESULTS: Annexin A2 gene and protein levels were down-regulated during in vitro decidualization of human endometrial stromal cells from women with previous severe preeclampsia compared with control individuals. To assess its role in the endometrial stroma, we inhibited Annexin A2 expression and detected decidualization failure as evidenced by impaired morphologic transformation, which was associated with altered actin polymerization and low prolactin and insulin-like growth binding protein-1 secretions. Functionally, in vitro models demonstrated that Annexin A2 inhibition failed to support embryo invasion. This finding was corroborated by reduced trophoblast spreading through human endometrial stromal cells, lack of motility of these cells, and reduced trophoblast invasion in the presence of conditioned media from Annexin A2-inhibited cells. Extending our discovery to an animal model, we detected that Annexin A2-null mice have a functional deficiency in decidualization and placentation that impairs fetal growth as a feature that is associated with severe preeclampsia. CONCLUSION: Together, in vitro and in vivo results suggest that endometrial defects in Annexin A2 expression impair decidualization of endometrial stromal cells as well as the uterine microenvironment that promotes embryo implantation and placentation. Our findings highlight the maternal contribution to the pathogenesis of severe preeclampsia and suggest that evaluation of Annexin A2 may provide a novel strategy to assess a woman's risk of experiencing this disease and perhaps discover therapeutic interventions to improve decidualization.


Assuntos
Anexina A2/genética , Anexina A2/metabolismo , Decídua/fisiopatologia , Pré-Eclâmpsia/genética , Actinas/metabolismo , Animais , Anexina A2/antagonistas & inibidores , Anexina A2/deficiência , Movimento Celular , Células Cultivadas , Decídua/patologia , Modelos Animais de Doenças , Implantação do Embrião , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Camundongos , Placentação/genética , Gravidez , Prolactina/metabolismo , RNA Interferente Pequeno/farmacologia , Células Estromais , Trofoblastos/fisiologia
5.
Am J Obstet Gynecol ; 221(4): 320.e1-320.e23, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31121144

RESUMO

BACKGROUND: Although uterine leiomyomas and leiomyosarcomas are considered biologically unrelated tumors, they share morphologic and histologic characteristics that complicate their differential diagnosis. The long-term therapeutic option for leiomyoma is laparoscopic myomectomy with morcellation, particularly for patients who wish to preserve their fertility. However, because of the potential dissemination of undiagnosed or hidden leiomyosarcoma from morcellation, there is a need to develop a preoperative assessment of malignancy risk. OBJECTIVE: Through an integrated comparative genomic and transcriptomic analysis, we aim to identify differential genetic targets in leiomyomas vs leiomyosarcomas using next-generation sequencing as the first step toward preoperative differential diagnosis. STUDY DESIGN: Targeted sequencing of DNA and RNA coding regions for solid tumor-associated genes was performed on formalin-fixed paraffin-embedded samples from 13 leiomyomas and 13 leiomyosarcoma cases. DNA sequencing was used to identify copy number variations, single-nucleotide variants, and small insertions/deletions. RNA sequencing was used to identify gene fusions, splice variants, and/or differential gene expression profiles. RESULTS: In leiomyosarcomas, tumor mutation burden was higher in terms of copy number variations, single nucleotide variants, small insertions/deletions, and gene fusions compared with leiomyomas. For copy number variations, 20 genes were affected by deletions in leiomyosarcomas, compared with 6 observed losses in leiomyomas. Gains (duplications) were identified in 19 genes in leiomyosarcomas, but only 3 genes in leiomyomas. The most common mutations (single-nucleotide variants and insertions/deletions) for leiomyosarcomas were identified in 105 genes of all analyzed leiomyosarcomas; 82 genes were affected in leiomyomas. Of note, 1 tumor previously diagnosed as leiomyosarcoma was established as inflammatory myofibroblastic tumor along this study with a novel ALK-TNS1 fusion. Finally, a differential transcriptomic profile was observed for 11 of 55 genes analyzed in leiomyosarcomas; 8.5% of initially diagnosed leiomyosarcomas showed high-confidence, novel gene fusions that were associated with these tumors. CONCLUSION: Through integrated comparative genomic and transcriptomic analyses, we identified novel differential genetic targets that potentially differentiate leiomyosarcomas and leiomyomas. This provides a new insight into the differential diagnosis of these myometrial tumors.


Assuntos
Leiomioma/diagnóstico , Leiomiossarcoma/diagnóstico , Neoplasias Uterinas/diagnóstico , Adulto , Idoso , Variações do Número de Cópias de DNA , Diagnóstico Diferencial , Feminino , Deleção de Genes , Duplicação Gênica , Perfilação da Expressão Gênica , Fusão Gênica , Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Leiomioma/genética , Leiomiossarcoma/genética , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA , Análise de Sequência de RNA , Neoplasias Uterinas/genética
7.
Proc Natl Acad Sci U S A ; 114(40): E8468-E8477, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28923940

RESUMO

In preeclampsia (PE), cytotrophoblast (CTB) invasion of the uterus and spiral arteries is often shallow. Thus, the placenta's role has been a focus. In this study, we tested the hypothesis that decidual defects are an important determinant of the placental phenotype. We isolated human endometrial stromal cells from nonpregnant donors with a previous pregnancy that was complicated by severe PE (sPE). Compared with control cells, they failed to decidualize in vitro as demonstrated by morphological criteria and the analysis of stage-specific antigens (i.e., IGFBP1, PRL). These results were bolstered by global transcriptional profiling data that showed they were transcriptionally inert. Additionally, we used laser microdissection to isolate the decidua from tissue sections of the maternal-fetal interface in sPE. Global transcriptional profiling revealed defects in gene expression. Also, decidual cells from patients with sPE, which dedifferentiated in vitro, failed to redecidualize in culture. Conditioned medium from these cells failed to support CTB invasion. To mimic aspects of the uterine environment in normal pregnancy, we added PRL and IGFBP1, which enhanced invasion. These data suggested that failed decidualization is an important contributor to down-regulated CTB invasion in sPE. Future studies will be aimed at determining whether this discovery has translational potential with regard to assessing a woman's risk of developing this pregnancy complication.


Assuntos
Decídua/patologia , Endométrio/patologia , Pré-Eclâmpsia/etiologia , Células Estromais/patologia , Trofoblastos/patologia , Adulto , Células Cultivadas , Decídua/metabolismo , Implantação do Embrião , Endométrio/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Pré-Eclâmpsia/patologia , Gravidez , Primeiro Trimestre da Gravidez , Células Estromais/metabolismo , Trofoblastos/metabolismo
8.
Fertil Steril ; 108(5): 832-842.e1, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28911927

RESUMO

OBJECTIVE: To characterize PGRMC1 and SERBP1 in human endometrium and to investigate the putative role of PGRMC1 in endometrial decidualization. DESIGN: The PGRMC1 and SERBP1 expression in human endometrium was determined throughout the menstrual cycle. We analyzed the colocalization of PGRMC1 and SERBP1. Then, endometrial stromal cells (ESCs) were isolated to investigate the functional effect of PGRMC1 overexpression on decidualization. SETTING: IVI clinic. PATIENT(S): Endometrial biopsies were collected from fertile volunteers (n = 61) attending the clinic as ovum donors. INTERVENTION(S): Endometrial samples of 61 healthy fertile women. MAIN OUTCOME MEASURE(S): In vivo localization of PGRMC1 and SERBP1 was assessed by immunohistochemistry. The PGRMC1/SERBP1 colocalization was investigated in vitro and in vivo. Decidualization effect of PGRMC1 overexpression was evaluated in primary ESC cultures. RESULT(S): The PGRMC1 was detected in the endometrial stroma throughout the menstrual cycle, but decreased in the late secretory phase. The SERBP1 immunostaining was present in stroma and increased in the entire the menstrual cycle. The PGRMC1 and SERBP1 colocalized in the cytoplasmic fractions of nondecidualized and decidualized ESC. The PGRMC1 overexpression significantly inhibited in vitro decidualization. CONCLUSION(S): Our results suggest that classic P receptors (PRs) are not the only kind playing a role in the normal physiology of the endometrium. The human decidualization process could be altered by the overexpression or mislocalization of PGRMC1 in ESC.


Assuntos
Decídua/metabolismo , Endométrio/metabolismo , Proteínas de Membrana/metabolismo , Ciclo Menstrual/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores de Progesterona/metabolismo , Células Estromais/metabolismo , Adolescente , Adulto , Células Cultivadas , Decídua/citologia , Endométrio/citologia , Feminino , Voluntários Saudáveis , Humanos , Fatores de Tempo , Adulto Jovem
9.
Am J Obstet Gynecol ; 217(2): 200.e1-200.e17, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28347715

RESUMO

BACKGROUND: The maternal signs of preeclampsia, which include the new onset of high blood pressure, can occur because of faulty placentation. We theorized that transcriptomic analyses of trophoblast subpopulations in situ would lend new insights into the role of these cells in preeclampsia pathogenesis. OBJECTIVE: Our goal was to enrich syncytiotrophoblasts, invasive cytotrophoblasts, or endovascular cytotrophoblasts from the placentas of severe preeclampsia cases. Total RNA was subjected to global transcriptional profiling to identify RNAs that were misexpressed compared with controls. STUDY DESIGN: This was a cross-sectional analysis of placentas from women who had been diagnosed with severe preeclampsia. Gestational age-matched controls were placentas from women who had a preterm birth with no signs of infection. Laser microdissection enabled enrichment of syncytiotrophoblasts, invasive cytotrophoblasts, or endovascular cytotrophoblasts. After RNA isolation, a microarray approach was used for global transcriptional profiling. Immunolocalization identified changes in messenger RNA expression that carried over to the protein level. Differential expression of non-protein-coding RNAs was confirmed by in situ hybridization. A 2-way analysis of variance of non-coding RNA expression identified particular classes that distinguished trophoblasts in cases vs controls. Cajal body foci were visualized by coilin immunolocalization. RESULTS: Comparison of the trophoblast subtype data within each group (severe preeclampsia or noninfected preterm birth) identified many highly differentially expressed genes. They included molecules that are known to be expressed by each subpopulation, which is evidence that the method worked. Genes that were expressed differentially between the 2 groups, in a cell-type-specific manner, encoded a combination of molecules that previous studies associated with severe preeclampsia and those that were not known to be dysregulated in this pregnancy complication. Gene ontology analysis of the syncytiotrophoblast data highlighted the dysregulation of immune functions, morphogenesis, transport, and responses to vascular endothelial growth factor and progesterone. The invasive cytotrophoblast data provided evidence of alterations in cellular movement, which is consistent with the shallow invasion often associated with severe preeclampsia. Other dysregulated pathways included immune, lipid, oxygen, and transforming growth factor-beta responses. The data for endovascular cytotrophoblasts showed disordered metabolism, signaling, and vascular development. Additionally, the transcriptional data revealed the differential expression in severe preeclampsia of 2 classes of non-coding RNAs: long non-coding RNAs and small nucleolar RNAs. The long non-coding RNA, urothelial cancer associated 1, was the most highly up-regulated in this class. In situ hybridization confirmed severe preeclampsia-associated expression in syncytiotrophoblasts. The small nucleolar RNAs, which chemically modify RNA structure, also correlated with severe preeclampsia. Thus, we enumerated Cajal body foci, sites of small nucleolar RNA activity, in primary cytotrophoblasts that were isolated from control and severe preeclampsia placentas. In severe preeclampsia, cytotrophoblasts had approximately double the number of these foci as the control samples. CONCLUSION: A laser microdissection approach enabled the identification of novel messenger RNAs and non-coding RNAs that were misexpressed by various trophoblast subpopulations in severe preeclampsia. The results suggested new avenues of investigation, in particular, the roles of PRG2, Kell blood group determinants, and urothelial cancer associated 1 in syncytiotrophoblast diseases. Additionally, many of the newly identified dysregulated molecules might have clinical utility as biomarkers of severe preeclampsia.


Assuntos
Pré-Eclâmpsia/genética , Pré-Eclâmpsia/patologia , Trofoblastos , Adulto , Estudos de Casos e Controles , Estudos Transversais , Feminino , Humanos , Gravidez , RNA Longo não Codificante/análise
10.
Development ; 144(5): 767-777, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28232601

RESUMO

Pre-eclampsia (PE), which affects ∼8% of first pregnancies, is associated with faulty placentation. Extravillous cytotrophoblasts (CTBs) fail to differentiate properly, contributing to shallow uterine invasion and deficient spiral artery remodeling. We studied the effects of severe PE (sPE) on the smooth chorion portion of the fetal membranes. The results showed a significant expansion of the CTB layer. The cells displayed enhanced expression of stage-specific antigens that extravillous CTBs normally upregulate as they exit the placenta. Transcriptomics revealed the dysregulated expression of many genes (e.g. placental proteins, markers of oxidative stress). We confirmed an sPE-related increase in production of PAPPA1, which releases IGF1 from its binding protein. IGF1 enhanced proliferation of smooth chorion CTBs, a possible explanation for expansion of this layer, which may partially compensate for the placental deficits.


Assuntos
Córion/metabolismo , Placenta/metabolismo , Placentação , Pré-Eclâmpsia/metabolismo , Trofoblastos/metabolismo , Adulto , Proliferação de Células , Córion/citologia , Membranas Extraembrionárias/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Queratinas/metabolismo , Estresse Oxidativo , Placenta/citologia , Pré-Eclâmpsia/patologia , Gravidez , Proteína Plasmática A Associada à Gravidez/metabolismo , Ligação Proteica , Transcrição Gênica , Transcriptoma , Trofoblastos/citologia
11.
Hum Reprod ; 29(9): 1957-67, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25106620

RESUMO

STUDY QUESTION: Are there any proteomic differences between receptive (R) and non-receptive (NR) endometrial receptivity array (ERA)-diagnosed endometria obtained on the same day of a hormonal replacement therapy (HRT) treatment cycle? SUMMARY ANSWER: There is a different proteomic signature between R and NR ERA-diagnosed endometrium obtained on the same day of HRT cycles. WHAT IS KNOWN ALREADY: The human endometrial transcriptome has been extensively investigated in the last decade resulting in the development of a new diagnostic test based on the transcriptomic signature of the window of implantation (WOI). Much less is known about the proteomics derived from the transcripts present during the WOI. STUDY DESIGN, SIZE, AND DURATION: This study was a basic proteomic analysis of human endometrial biopsies taken from twelve IVF patients. PARTICIPANTS/MATERIALS, SETTING, AND METHODS: Human endometrial biopsies were collected during HRT cycles after 5 days of progesterone (P) administration, and diagnosed as receptive (R; n = 6) or non-receptive (NR; n = 6) by the ERA test. Endometrial proteins were extracted, labelled and separated using differential in-gel electrophoresis (DIGE). Proteins were identified using mass spectrometry, followed up by in silico analysis. Validation studies using western blots and immunolocalization were performed for the progesterone receptor membrane component 1 (PGRMC1) and annexin A6 (ANXA6) proteins. MAIN RESULTS AND THE ROLE OF CHANCE: DIGE analysis followed by protein identification by MALDI-MS and database searches revealed 24 differentially expressed proteins in R versus NR samples. In silico analysis showed two pathways which were significantly different between R and NR samples. Expression of PGRMC1 and ANXA6 was validated and localized by western blots and immunohistochemistry. These results highlight these proteins as key targets likely to be important in the comprehension of human endometrial receptivity. LIMITATIONS, REASONS FOR CAUTION: This was mainly a descriptive study with no functional studies on the proteins found. We also used a low number of human endometrial samples for the DIGE analysis. WIDER IMPLICATIONS OF THE FINDINGS: This study identified the proteomic profile associated with receptive or non-receptive human endometria. Our findings suggest that although histological dating indicates a putative 'receptive' status within the WOI, a different transcriptomic and proteomic profile is observed in these samples. We should move towards using more personalized WOIs, where identification of the correct endometrial receptivity status, and consequently the success of IVF, relies on individual molecular signatures rather than traditional endometrial dating. STUDY FUNDING/COMPETING INTERESTS: F.D.'s participation in this work was supported by the Spanish Ministry of Economy and Competitiveness, through the Miguel Servet Programme (CP13/00075) co-founded by FEDER. The project was also supported by a grant from the Spanish Ministry of Economy and Competitiveness, through the FIS Programme (PI12/00450). The authors have no financial/commercial conflicts of interest to declare.


Assuntos
Implantação do Embrião , Endométrio/fisiologia , Proteoma , Anexina A6/análise , Anexina A6/metabolismo , Endométrio/metabolismo , Feminino , Fertilização in vitro , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Espectrometria de Massas , Proteínas de Membrana/análise , Proteínas de Membrana/metabolismo , Progesterona/uso terapêutico , Proteômica , Receptores de Progesterona/análise , Receptores de Progesterona/metabolismo
12.
Fertil Steril ; 99(4): 1078-85, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23312228

RESUMO

This article highlights the need for methods to objectively diagnose endometrial receptivity as a factor contributing to infertility in female patients. The correct identification of the appropriate window of implantation in a given patient, by using endometrial receptivity biomarkers, can help to prevent reproductive failure resulting from misplaced timing of the endometrial window of implantation (WOI). Although to date no single, clinically relevant morphologic, molecular, or histologic marker capable of indicating endometrial receptivity status has been identified, global transcriptomic analysis of human endometria performed in the last decade has given us insights into a genomic signature that is capable of identifying endometrial receptivity. As a consequence, a genomic tool named the Endometrial Receptivity Array (ERA), based on a customized microarray, was developed, and along with it a specially trained bioinformatic prediction computer algorithm was created to identify WOI timing in the endometrium. This tool has proven more accurate and consistent than histologic (Noyes) dating at identifying the personalized WOI day, thus leading to the new clinical concept of personalized ET on the optimum day of endometrial receptivity, identified individually case by case.


Assuntos
Transferência Embrionária/métodos , Endométrio/fisiologia , Fertilização in vitro/métodos , Infertilidade Feminina/genética , Transcriptoma , Biomarcadores/metabolismo , Feminino , Humanos , Infertilidade Feminina/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez
13.
FASEB J ; 26(9): 3715-27, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22645245

RESUMO

Annexin A2 (ANXA2) is present in vivo in the mid- and late-secretory endometria and is mainly localized in the luminal epithelium. Our aim was to evaluate its function in regulating the human implantation process. With an in vitro adhesion model, constructed to evaluate how the mouse embryo and JEG-3 spheroids attach to human endometrial epithelial cells, we demonstrated that ANXA2 inhibition significantly diminishes embryo adhesiveness. ANXA2 is also implicated in endometrial epithelial cell migration and trophoblast outgrowth. ANXA2 was seen to be linked to the RhoA/ROCK pathway and to regulate cell adhesion. We noted that ANXA2 inhibition significantly reduces active RhoA, although RhoA inactivation does not alter the ANXA2 levels. RhoA inactivation and ROCK inhibition also moderate embryo adhesiveness to endometrial epithelial cells. We corroborated that the induction of constitutively active RhoA partially reverses the effects of ANXA2 inhibition on endometrial adhesiveness. These molecules colocalize on the plasma membrane of endometrial epithelial cells, and a large proportion of ANXA2 and RhoA are colocalized in the F-actin networks. The functional effects of ANXA2 inhibition and RhoA/ROCK inactivation are associated with significant alterations in F-actin organization and its depolymerization. ANXA2 may act upstream of the RhoA/ROCK pathway by regulating F-actin remodeling and is a key factor in human endometrial adhesiveness.


Assuntos
Actinas/fisiologia , Anexina A2/fisiologia , Implantação do Embrião , Endométrio/citologia , Proteína rhoA de Ligação ao GTP/fisiologia , Adolescente , Adulto , Anexina A2/antagonistas & inibidores , Sequência de Bases , Células Cultivadas , Primers do DNA , Feminino , Humanos , RNA Interferente Pequeno , Regulação para Cima , Adulto Jovem
14.
J Clin Endocrinol Metab ; 96(3): 706-16, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21190976

RESUMO

CONTEXT: Decidualization of the human endometrium, which involves morphological and biochemical modifications of the endometrial stromal cells (ESCs), is a prerequisite for adequate trophoblast invasion and placenta formation. OBJECTIVE: This study aims to investigate the proteome and secretome of in vitro decidualized ESCs. These data were combined with published genomic information and integrated to model the human decidualization interactome. DESIGN: Prospective experimental case-control study. SETTING: A private research foundation. PATIENTS: Sixteen healthy volunteer ovum donors. INTERVENTION: Endometrial samples were obtained, and ESCs were isolated and decidualized in vitro. MAIN OUTCOME MEASURES: Two-dimensional difference in-gel electrophoresis, matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry, Western blot, human protein cytokine array, ELISA, and bioinformatics analysis were performed. RESULTS: The proteomic analysis revealed 60 differentially expressed proteins (36 over- and 24 underexpressed) in decidualized versus control ESCs, including known decidualization markers (cathepsin B) and new biomarkers (transglutaminase 2, peroxiredoxin 4, and the ACTB protein). In the secretomic analysis, a total of 13 secreted proteins (11 up- and 2 down-regulated) were identified, including well-recognized markers (IGF binding protein-1 and prolactin) and novel ones (myeloid progenitor inhibitory factor-1 and platelet endothelial cell adhesion molecule-1). These proteome/secretome profiles have been integrated into a decidualization interactome model. CONCLUSIONS: Proteomic and secretomic have been used as hypothesis-free approaches together with complex bioinformatics to model the human decidual interactome for the first time. We confirm previous knowledge, describe new molecules, and we have built up a model for human in vitro decidualization as invaluable tool for the diagnosis, therapy, and interpretation of biological phenomena.


Assuntos
Decídua/fisiologia , Endométrio/fisiologia , Proteoma/genética , Adolescente , Adulto , Biomarcadores/análise , Western Blotting , Estudos de Casos e Controles , Forma Celular , Células Cultivadas , Biologia Computacional , Meios de Cultivo Condicionados , Citocinas/metabolismo , Decídua/citologia , Eletroforese em Gel de Poliacrilamida , Endométrio/citologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Modelos Biológicos , Processamento de Proteína Pós-Traducional , Proteômica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Células Estromais/fisiologia , Células Estromais/ultraestrutura , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...