Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Hum Reprod ; 37(4): 747-761, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35092277

RESUMO

STUDY QUESTION: Can the accuracy of timing of luteal phase endometrial biopsies based on urinary ovulation testing be improved by measuring the expression of a small number of genes and a continuous, non-categorical modelling approach? SUMMARY ANSWER: Measuring the expression levels of six genes (IL2RB, IGFBP1, CXCL14, DPP4, GPX3 and SLC15A2) is sufficient to obtain substantially more accurate timing estimates and to assess the reliability of timing estimates for each sample. WHAT IS KNOWN ALREADY: Commercially available endometrial timing approaches based on gene expression require large gene sets and use a categorical approach that classifies samples as pre-receptive, receptive or post-receptive. STUDY DESIGN, SIZE, DURATION: Gene expression was measured by RTq-PCR in different sample sets, comprising a total of 664 endometrial biopsies obtained 4-12 days after a self-reported positive home ovulation test. A further 36 endometrial samples were profiled by RTq-PCR as well as RNA-sequencing. PARTICIPANTS/MATERIALS, SETTING, METHODS: A computational procedure, named 'EndoTime', was established that models the temporal profile of each gene and estimates the timing of each sample. Iterating these steps, temporal profiles are gradually refined as sample timings are being updated, and confidence in timing estimates is increased. After convergence, the method reports updated timing estimates for each sample while preserving the overall distribution of time points. MAIN RESULTS AND THE ROLE OF CHANCE: The Wilcoxon rank-sum test was used to confirm that ordering samples by EndoTime estimates yields sharper temporal expression profiles for held-out genes (not used when determining sample timings) than ordering the same expression values by patient-reported times (GPX3: P < 0.005; CXCL14: P < 2.7e-6; DPP4: P < 3.7e-13). Pearson correlation between EndoTime estimates for the same sample set but based on RTq-PCR or RNA-sequencing data showed a high degree of congruency between the two (P = 8.6e-10, R2 = 0.687). Estimated timings did not differ significantly between control subjects and patients with recurrent pregnancy loss or recurrent implantation failure (P > 0.05). LARGE SCALE DATA: The RTq-PCR data files are available via the GitHub repository for the EndoTime software at https://github.com/AE-Mitchell/EndoTime, as is the code used for pre-processing of RTq-PCR data. The RNA-sequencing data are available on GEO (accession GSE180485). LIMITATIONS, REASONS FOR CAUTION: Timing estimates are informed by glandular gene expression and will only represent the temporal state of other endometrial cell types if in synchrony with the epithelium. Methods that estimate the day of ovulation are still required as these data are essential inputs in our method. Our approach, in its current iteration, performs batch correction such that larger sample batches impart greater accuracy to timing estimations. In theory, our method requires endometrial samples obtained at different days in the luteal phase. In practice, however, this is not a concern as timings based on urinary ovulation testing are associated with a sufficient level of noise to ensure that a variety of time points will be sampled. WIDER IMPLICATIONS OF THE FINDINGS: Our method is the first to assay the temporal state of luteal-phase endometrial samples on a continuous domain. It is freely available with fully shared data and open-source software. EndoTime enables accurate temporal profiling of any gene in luteal endometrial samples for a wide range of research applications and, potentially, clinical use. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by a Wellcome Trust Investigator Award (Grant/Award Number: 212233/Z/18/Z) and the Tommy's National Miscarriage Research Centre. None of the authors have any competing interests. J.L. was funded by the Biotechnology and Biological Sciences Research Council (UK) through the Midlands Integrative Biology Training Partnership (MIBTP, BB/M01116X/1).


Assuntos
Aborto Habitual , Endométrio , Aborto Habitual/metabolismo , Endométrio/metabolismo , Feminino , Humanos , Fase Luteal/metabolismo , Gravidez , Reprodutibilidade dos Testes , Análise de Sequência de RNA
2.
Stem Cells ; 39(8): 1067-1080, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33764639

RESUMO

Pregnancy depends on the wholesale transformation of the endometrium, a process driven by differentiation of endometrial stromal cells (EnSC) into specialist decidual cells. Upon embryo implantation, decidual cells impart the tissue plasticity needed to accommodate a rapidly growing conceptus and invading placenta, although the underlying mechanisms are unclear. Here we characterize a discrete population of highly proliferative mesenchymal cells (hPMC) in midluteal human endometrium, coinciding with the window of embryo implantation. Single-cell transcriptomics demonstrated that hPMC express genes involved in chemotaxis and vascular transmigration. Although distinct from resident EnSC, hPMC also express genes encoding pivotal decidual transcription factors and markers, most prominently prolactin. We further show that hPMC are enriched around spiral arterioles, scattered throughout the stroma, and occasionally present in glandular and luminal epithelium. The abundance of hPMC correlated with the in vitro colony-forming unit activity of midluteal endometrium and, conversely, clonogenic cells in culture express a gene signature partially conserved in hPMC. Cross-referencing of single-cell RNA-sequencing data sets indicated that hPMC differentiate into a recently discovered decidual subpopulation in early pregnancy. Finally, we demonstrate that recurrent pregnancy loss is associated with hPMC depletion. Collectively, our findings characterize midluteal hPMC as novel decidual precursors that are likely derived from circulating bone marrow-derived mesenchymal stem/stromal cells and integral to decidual plasticity in pregnancy.


Assuntos
Implantação do Embrião , Endométrio , Diferenciação Celular , Decídua , Embrião de Mamíferos , Feminino , Humanos , Gravidez , Células Estromais
3.
J Obstet Gynaecol Res ; 47(2): 689-697, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33274569

RESUMO

OBJECTIVE: To determine the value of uterine CD138+ cells, as a marker of chronic endometritis, in predicting subsequent reproductive outcome in women with history of recurrent pregnancy loss. DESIGN: A prospective longitudinal study. SETTING: Tertiary specialized clinic. PATIENTS: Women with history of recurrent pregnancy loss or implantation failure over a 12-months follow-up period. INTERVENTION: We quantified the CD138+ cells/high powered field (hpf) using immunohistochemistry and image analysis of endometrial biopsies obtained during the secretory stage post ovulation. MAIN OUTCOME MEASURES: Live birth and subsequent pregnancy loss. We calculated the receiver operator curve for predicting subsequent pregnancy loss and reported using relative risk (RR) and 95% confidence intervals (CI). RESULTS: We enrolled 344 women of whom 88 became pregnant (88/344, 25.5%). Half of them had a subsequent live birth (47/88, 53%) and the rest lost their pregnancy (41/88, 46%). The median CD138+ score was significantly lower in the live birth group (P < 0.005) and women with a CD138+ score ≥ 16/hpf had a higher risk of subsequent miscarriage (RR 10.0, 95% CI 2.78-36.02). CD138+ cells count showed a good prediction for subsequent pregnancy loss in high-risk women with an area under the curve of 0.75 (95% CI 0.59-0.82, P = 0.01). A cut-off value of 4-6 cells/hpf offered the best predictive accuracy with higher scores predicting worse reproductive outcome. Our findings are limited by the small event rate and the sample size of our cohort. CONCLUSION: Quantifying CD138+ cells by immunohistochemistry in women with a history of recurrent pregnancy loss is helpful to diagnose chronic endometritis and predict subsequent reproductive outcome.


Assuntos
Aborto Habitual , Endometrite , Estudos de Coortes , Endometrite/diagnóstico , Endometrite/epidemiologia , Endométrio , Feminino , Humanos , Estudos Longitudinais , Gravidez , Estudos Prospectivos
4.
Reproduction ; 152(5): R159-66, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27430234

RESUMO

Endometrial stem-like cells, including mesenchymal stem cells (MSCs) and epithelial progenitor cells, are essential for cyclic regeneration of the endometrium following menstrual shedding. Emerging evidence indicates that endometrial MSCs (eMSCs) constitute a dynamic population of cells that enables the endometrium to adapt in response to a failed pregnancy. Recurrent miscarriage is associated with relative depletion of endometrial eMSCs, which not only curtails the intrinsic ability of the endometrium to adapt to reproductive failure but also compromises endometrial decidualization, an obligatory transformation process for embryo implantation. These novel findings should pave the way for more effective screening of women at risk of pregnancy failure before conception.


Assuntos
Aborto Habitual/prevenção & controle , Implantação do Embrião , Endométrio/citologia , Células-Tronco Mesenquimais/citologia , Regeneração/fisiologia , Aborto Habitual/fisiopatologia , Feminino , Humanos , Gravidez
5.
Dev Biol ; 338(2): 215-25, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20004186

RESUMO

Regulated neuron production within the vertebrate nervous system relies on input from multiple signalling pathways. Work in the Drosophila retina has demonstrated that PI3-kinase and downstream TOR signalling regulate the timing of photoreceptor differentiation; however, the function of such signals during vertebrate neurogenesis is not well understood. Here we show that mutant mice lacking PKB activity downstream of PDK1, the master kinase of the PI3-kinase pathway, exhibit deficient neuron production. We further demonstrate expression of PI3-kinase signalling components and active PKB and TOR signalling in the chick spinal cord, an early site of neurogenesis. Neuron production was also attenuated in the chick neural tube following exposure to small molecule inhibitors of PI3-kinase (LY294002) or TOR (Rapamycin) activity. Furthermore, Rapamycin repressed expression of early neuronal differentiation genes, such as Ngn2, but did not inhibit expression of Sox1B genes characteristic of proliferating neural progenitors. In addition, some cells expressing an early neuronal marker were mis-localised at the ventricular surface in the presence of Rapamycin and remained aberrantly within the cell cycle. These findings suggest that TOR signalling is necessary to initiate neuronal differentiation and that it may facilitate coordination of cell cycle and differentiation programmes. In contrast, stimulating PI3-kinase signalling did not increase neuron production, suggesting that such activity is simply permissive for vertebrate neurogenesis.


Assuntos
Diferenciação Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Tubo Neural/citologia , Neurônios/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Animais , Ciclo Celular , Diferenciação Celular/genética , Embrião de Galinha , Camundongos , Camundongos Mutantes , Tubo Neural/metabolismo , Neurogênese , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
6.
Elife ; 102021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34487490

RESUMO

Decidual remodelling of midluteal endometrium leads to a short implantation window after which the uterine mucosa either breaks down or is transformed into a robust matrix that accommodates the placenta throughout pregnancy. To gain insights into the underlying mechanisms, we established and characterized endometrial assembloids, consisting of gland-like organoids and primary stromal cells. Single-cell transcriptomics revealed that decidualized assembloids closely resemble midluteal endometrium, harbouring differentiated and senescent subpopulations in both glands and stroma. We show that acute senescence in glandular epithelium drives secretion of multiple canonical implantation factors, whereas in the stroma it calibrates the emergence of anti-inflammatory decidual cells and pro-inflammatory senescent decidual cells. Pharmacological inhibition of stress responses in pre-decidual cells accelerated decidualization by eliminating the emergence of senescent decidual cells. In co-culture experiments, accelerated decidualization resulted in entrapment of collapsed human blastocysts in a robust, static decidual matrix. By contrast, the presence of senescent decidual cells created a dynamic implantation environment, enabling embryo expansion and attachment, although their persistence led to gradual disintegration of assembloids. Our findings suggest that decidual senescence controls endometrial fate decisions at implantation and highlight how endometrial assembloids may accelerate the discovery of new treatments to prevent reproductive failure.


At the beginning of a human pregnancy, the embryo implants into the uterus lining, known as the endometrium. At this point, the endometrium transforms into a new tissue that helps the placenta to form. Problems in this transformation process are linked to pregnancy disorders, many of which can lead to implantation failure (the embryo fails to invade the endometrium altogether) or recurrent miscarriages (the embryo implants successfully, but the interface between the placenta and the endometrium subsequently breaks down). Studying the implantation of human embryos directly is difficult due to ethical and technical barriers, and animals do not perfectly mimic the human process, making it challenging to determine the causes of pregnancy disorders. However, it is likely that a form of cellular arrest called senescence, in which cells stop dividing but remain metabolically active, plays a role. Indeed, excessive senescence in the cells that make up the endometrium is associated with recurrent miscarriage, while a lack of senescence is associated with implantation failure. To study this process, Rawlings et al. developed a new laboratory model of the human endometrium by assembling two of the main cell types found in the tissue into a three-dimensional structure. When treated with hormones, these 'assembloids' successfully mimic the activity of genes in the cells of the endometrium during implantation. Rawlings et al. then exposed the assembloids to the drug dasatinib, which targets and eliminates senescent cells. This experiment showed that assembloids become very robust and static when devoid of senescent cells. Rawlings et al. then studied the interaction between embryos and assembloids using time-lapse imaging. In the absence of dasatinib treatment, cells in the assembloid migrated towards the embryo as it expanded, a process required for implantation. However, when senescent cells were eliminated using dasatinib, this movement of cells towards the embryo stopped, and the embryo failed to expand, in a situation that mimicks implantation failure. The assembloid model of the endometrium may help scientists to study endometrial defects in the lab and test potential treatments. Further work will include other endometrial cell types in the assembloids, and could help increase the reliability of the model. However, any drug treatments identified using this model will need further research into their safety and effectiveness before they can be offered to patients.


Assuntos
Senescência Celular , Implantação do Embrião/fisiologia , Endométrio/citologia , Células Estromais/citologia , Técnicas de Cocultura , Decídua/fisiologia , Feminino , Humanos , Organoides , Gravidez
7.
Front Cell Dev Biol ; 8: 621016, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33537312

RESUMO

Vascular adhesion protein-1 (VAP-1) is an inflammation-inducible adhesion molecule and a primary amine oxidase involved in immune cell trafficking. Leukocyte extravasation into tissues is mediated by adhesion molecules expressed on endothelial cells and pericytes. Pericytes play a major role in the angiogenesis and vascularization of cycling endometrium. However, the functional properties of pericytes in the human endometrium are not known. Here we show that pericytes surrounding the spiral arterioles in midluteal human endometrium constitutively express VAP-1. We first characterize these pericytes and demonstrate that knockdown of VAP-1 perturbed their biophysical properties and compromised their contractile, migratory, adhesive and clonogenic capacities. Furthermore, we show that loss of VAP-1 disrupts pericyte-uterine natural killer cell interactions in vitro. Taken together, the data not only reveal that endometrial pericytes represent a cell population with distinct biophysical and functional properties but also suggest a pivotal role for VAP-1 in regulating the recruitment of innate immune cells in human endometrium. We posit that VAP-1 could serve as a potential biomarker for pregnancy pathologies caused by a compromised perivascular environment prior to conception.

8.
Commun Biol ; 3(1): 37, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31965050

RESUMO

During the implantation window, the endometrium becomes poised to transition to a pregnant state, a process driven by differentiation of stromal cells into decidual cells (DC). Perturbations in this process, termed decidualization, leads to breakdown of the feto-maternal interface and miscarriage, but the underlying mechanisms are poorly understood. Here, we reconstructed the decidual pathway at single-cell level in vitro and demonstrate that stromal cells first mount an acute stress response before emerging as DC or senescent DC (snDC). In the absence of immune cell-mediated clearance of snDC, secondary senescence transforms DC into progesterone-resistant cells that abundantly express extracellular matrix remodelling factors. Additional single-cell analysis of midluteal endometrium identified DIO2 and SCARA5 as marker genes of a diverging decidual response in vivo. Finally, we report a conspicuous link between a pro-senescent decidual response in peri-implantation endometrium and recurrent pregnancy loss, suggesting that pre-pregnancy screening and intervention may reduce the burden of miscarriage.


Assuntos
Aborto Habitual/etiologia , Senescência Celular , Decídua/metabolismo , Implantação do Embrião , Aborto Habitual/metabolismo , Linhagem Celular , Senescência Celular/genética , Suscetibilidade a Doenças , Implantação do Embrião/genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Humanos , Vigilância Imunológica , Modelos Biológicos , Gravidez , Transdução de Sinais , Análise de Célula Única , Transcriptoma
9.
EBioMedicine ; 51: 102597, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31928963

RESUMO

BACKGROUND: Recurrent pregnancy loss (RPL) is associated with the loss of endometrial mesenchymal stem-like progenitor cells (eMSC). DPP4 inhibitors may increase homing and engraftment of bone marrow-derived cells to sites of tissue injury. Here, we evaluated the effect of the DPP4 inhibitor sitagliptin on eMSC in women with RPL, determined the impact on endometrial decidualization, and assessed the feasibility of a full-scale clinical trial. METHODS: A double-blind, randomised, placebo-controlled feasibility trial on women aged 18 to 42 years with a history of 3 or more miscarriages, regular menstrual cycles, and no contraindications to sitagliptin. Thirty-eight subjects were randomised to either 100 mg sitagliptin daily for 3 consecutive cycles or identical placebo capsules. Computer generated, permuted block randomisation was used to allocate treatment packs. Colony forming unit (CFU) assays were used to quantify eMSC in midluteal endometrial biopsies. The primary outcome measure was CFU counts. Secondary outcome measures were endometrial thickness, study acceptability, and first pregnancy outcome within 12 months following the study. Tissue samples were subjected to explorative investigations. FINDINGS: CFU counts following sitagliptin were higher compared to placebo only when adjusted for baseline CFU counts and age (RR: 1.52, 95% CI: 1.32-1.75, P<0.01). The change in CFU count was 1.68 in the sitagliptin group and 1.08 in the placebo group. Trial recruitment, acceptability, and drug compliance were high. There were no serious adverse events. Explorative investigations showed that sitagliptin inhibits the expression of DIO2, a marker gene of senescent decidual cells. INTERPRETATION: Sitagliptin increases eMSCs and decreases decidual senescence. A large-scale clinical trial evaluating the impact of preconception sitagliptin treatment on pregnancy outcome in RPL is feasible and warranted. FUNDING: Tommy's Baby Charity. CLINICAL TRIAL REGISTRATION: EU Clinical Trials Register no. 2016-001120-54.


Assuntos
Endométrio/citologia , Células-Tronco Mesenquimais/citologia , Fosfato de Sitagliptina/farmacologia , Administração Oral , Adulto , Ensaio de Unidades Formadoras de Colônias , Dipeptidil Peptidase 4/metabolismo , Método Duplo-Cego , Estudos de Viabilidade , Feminino , Humanos , Seleção de Pacientes , Placebos , Gravidez , Resultado da Gravidez , Análise de Regressão , Fosfato de Sitagliptina/administração & dosagem
10.
Elife ; 62017 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-29227245

RESUMO

In cycling human endometrium, menstruation is followed by rapid estrogen-dependent growth. Upon ovulation, progesterone and rising cellular cAMP levels activate the transcription factor Forkhead box O1 (FOXO1) in endometrial stromal cells (EnSCs), leading to cell cycle exit and differentiation into decidual cells that control embryo implantation. Here we show that FOXO1 also causes acute senescence of a subpopulation of decidualizing EnSCs in an IL-8 dependent manner. Selective depletion or enrichment of this subpopulation revealed that decidual senescence drives the transient inflammatory response associated with endometrial receptivity. Further, senescent cells prevent differentiation of endometrial mesenchymal stem cells in decidualizing cultures. As the cycle progresses, IL-15 activated uterine natural killer (uNK) cells selectively target and clear senescent decidual cells through granule exocytosis. Our findings reveal that acute decidual senescence governs endometrial rejuvenation and remodeling at embryo implantation, and suggest a critical role for uNK cells in maintaining homeostasis in cycling endometrium.


Assuntos
Senescência Celular , Decídua/citologia , Endométrio/citologia , Células Matadoras Naturais/citologia , Células Estromais/citologia , Útero/citologia , Diferenciação Celular , Células Cultivadas , Decídua/metabolismo , Endométrio/metabolismo , Feminino , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica , Humanos , Interleucina-15/metabolismo , Interleucina-8/metabolismo , Células Matadoras Naturais/metabolismo , Transdução de Sinais , Células Estromais/metabolismo , Útero/metabolismo
11.
PLoS One ; 10(7): e0132934, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26161852

RESUMO

Studies in humans and animal models show that neuronal insulin resistance increases the risk of developing Alzheimer's Disease (AD), and that insulin treatment may promote memory function. Cholinergic neurons play a critical role in cognitive and attentional processing and their dysfunction early in AD pathology may promote the progression of AD pathology. Synthesis and release of the neurotransmitter acetylcholine (ACh) is closely linked to the activity of the high-affinity choline transporter protein (CHT), but the impact of insulin receptor signaling and neuronal insulin resistance on these aspects of cholinergic function are unknown. In this study, we used differentiated SH-SY5Y cells stably-expressing CHT proteins to study the effect of insulin signaling on CHT activity and function. We find that choline uptake activity measured after acute addition of 20 nM insulin is significantly lower in cells that were grown for 24 h in media containing insulin compared to cells grown in the absence of insulin. This coincides with loss of ability to increase phospho-Protein Kinase B (PKB)/Akt levels in response to acute insulin stimulation in the chronic insulin-treated cells. Inhibition of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3-kinase) in cells significantly lowers phospho-PKB/Akt levels and decreases choline uptake activity. We show total internal reflection microscopy (TIRF) imaging of the dynamic movement of CHT proteins in live cells in response to depolarization and drug treatments. These data show that acute exposure of depolarized cells to insulin is coupled to transiently increased levels of CHT proteins at the cell surface, and that this is attenuated by chronic insulin exposure. Moreover, prolonged inhibition of PI3-kinase results in enhanced levels of CHT proteins at the cell surface by decreasing their rate of internalization.


Assuntos
Insulina/farmacologia , Proteínas de Membrana Transportadoras/metabolismo , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Colina/metabolismo , Cromonas/farmacologia , Endocitose/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Potenciais da Membrana/efeitos dos fármacos , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
12.
Mech Dev ; 129(9-12): 275-83, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22771518

RESUMO

The neural crest is a population of migratory cells that follows specific pathways during development, eventually differentiating to form parts of the face, heart, and peripheral nervous system, the latter of which includes contributions from placodal cells derived from the ectoderm. Stationary, premigratory neural crest cells acquire the capacity to migrate by undergoing an epithelial-to-mesenchymal transition that facilitates their emigration from the dorsal neural tube. This emigration involves, in part, the dismantling of cell-cell junctions, including apically localized tight junctions in the neuroepithelium. In this study, we have characterized the role of the transmembrane tight junction protein claudin-1 during neural crest and placode ontogeny. Our data indicate that claudin-1 is highly expressed in the developing neuroepithelium but is down-regulated in migratory neural crest cells, although expression persists in the ectoderm from which the placode cells arise. Depletion or overexpression of claudin-1 augments or reduces neural crest cell emigration, respectively, but does not impact the development of several cranial placodes. Taken together, our results reveal a novel function for a tight junction protein in the formation of migratory cranial neural crest cells in the developing vertebrate embryo.


Assuntos
Movimento Celular/fisiologia , Claudina-1/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Crista Neural/fisiologia , Animais , Movimento Celular/genética , Embrião de Galinha , Galinhas , Claudina-1/biossíntese , Claudina-1/genética , Claudina-1/metabolismo , Regulação para Baixo , Ectoderma/crescimento & desenvolvimento , Ectoderma/metabolismo , Epitélio/metabolismo , Crista Neural/metabolismo , Tubo Neural/metabolismo , Tubo Neural/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Neurônios/fisiologia , Junções Íntimas/genética , Junções Íntimas/metabolismo
13.
Cell Stem Cell ; 8(5): 538-51, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21549328

RESUMO

The transcription factor SOX2 is widely known to play a critical role in the central nervous system; however, its role in peripheral neurogenesis remains poorly understood. We recently developed an hESC-based model in which migratory cells undergo epithelial to mesenchymal transition (EMT) to acquire properties of neural crest (NC) cells. In this model, we found that migratory NC progenitors downregulate SOX2, but then start re-expressing SOX2 as they differentiate to form neurogenic dorsal root ganglion (DRG)-like clusters. SOX2 downregulation was sufficient to induce EMT and resulted in massive apoptosis when neuronal differentiation was induced. In vivo, downregulation of SOX2 in chick and mouse NC cells significantly reduced the numbers of neurons within DRG. We found that SOX2 binds directly to NGN1 and MASH1 promoters and is required for their expression. Our data suggest that SOX2 plays a key role for NGN1-dependent acquisition of neuronal fates in sensory ganglia.


Assuntos
Células-Tronco Embrionárias/metabolismo , Gânglios Espinais/metabolismo , Neurogênese , Fatores de Transcrição SOXB1/metabolismo , Células Receptoras Sensoriais/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Contagem de Células , Movimento Celular , Galinhas , Células-Tronco Embrionárias/citologia , Transição Epitelial-Mesenquimal , Gânglios Espinais/citologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Crista Neural/citologia , Neurogênese/genética , Especificidade de Órgãos , Ligação Proteica , Fatores de Transcrição SOXB1/genética
14.
J Clin Invest ; 119(6): 1438-49, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19487820

RESUMO

The events that convert adherent epithelial cells into individual migratory cells that can invade the extracellular matrix are known collectively as epithelial-mesenchymal transition (EMT). Throughout evolution, the capacity of cells to switch between these two cellular states has been fundamental in the generation of complex body patterns. Here, we review the EMT events that build the embryo and further discuss two prototypical processes governed by EMT in amniotes: gastrulation and neural crest formation. Cells undergo EMT to migrate and colonize distant territories. Not surprisingly, this is also the mechanism used by cancer cells to disperse throughout the body.


Assuntos
Diferenciação Celular , Células Epiteliais/citologia , Células-Tronco Mesenquimais/citologia , Animais , Movimento Celular , Doença , Humanos
15.
Dev Biol ; 302(2): 536-52, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17123506

RESUMO

During early vertebrate development Fibroblast Growth Factor (FGF) signalling is required for multiple activities including specification of mesodermal, neural and heart tissue, as well as gastrulation movements and regulation of differentiation and pattern onset in the extending body axis. A current challenge is to understand how FGF signalling generates such diverse outcomes. A key FGF downstream pathway is the Ras-MAPK/Erk1/2 cascade, which culminates in the phosphorylation of target proteins, such as the Ets family of transcription factors. To begin to assess specificity downstream of FGF in the chick embryo we have characterised the patterns of Fgfr1-4 expression and Erk1/2 activation, as well as expression of the Erk1/2 specific phosphatase, Mkp3 and of three Ets factor genes (Erm, Pea3 and Er81) from early blastula to the 10 somite stage. We identify new sites of Fgfr expression and show that nearly all regions of Erk1/2 activity are within Fgfr expression domains and require FGF signalling. Differences in intensity, duration, distribution and sub-cellular localisation of activated Erk1/2 are observed in distinct cell populations within the embryo and during wound healing. With few exceptions, a tight correspondence between Erk1/2 activation and Mkp3 expression is found, while specific combinations of Ets factors are associated with distinct regions of Erk1/2 activation. These findings provide a comprehensive spatial and temporal map of FGF/Erk1/2 activity during early chick development and identify region and tissue specific differences in expression of Fgfrs as well as Erk1/2 phosphorylation and transcriptional targets which help to define response specificity.


Assuntos
Fatores de Crescimento de Fibroblastos/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Animais , Embrião de Galinha , Fosfatase 6 de Especificidade Dupla , Ativação Enzimática , Regulação da Expressão Gênica no Desenvolvimento , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/biossíntese , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/biossíntese , Transdução de Sinais , Fatores de Transcrição/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA