Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell Mol Life Sci ; 81(1): 270, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38886218

RESUMEN

Early trophoblast differentiation is crucial for embryo implantation, placentation and fetal development. Dynamic changes in DNA methylation occur during preimplantation development and are critical for cell fate determination. However, the underlying regulatory mechanism remains unclear. Recently, we derived morula-like expanded potential stem cells from human preimplantation embryos (hEPSC-em), providing a valuable tool for studying early trophoblast differentiation. Data analysis on published datasets showed differential expressions of DNA methylation enzymes during early trophoblast differentiation in human embryos and hEPSC-em derived trophoblastic spheroids. We demonstrated downregulation of DNA methyltransferase 3 members (DNMT3s) and upregulation of ten-eleven translocation methylcytosine dioxygenases (TETs) during trophoblast differentiation. While DNMT inhibitor promoted trophoblast differentiation, TET inhibitor hindered the process and reduced implantation potential of trophoblastic spheroids. Further integrative analysis identified that glutamyl aminopeptidase (ENPEP), a trophectoderm progenitor marker, was hypomethylated and highly expressed in trophoblast lineages. Concordantly, progressive loss of DNA methylation in ENPEP promoter and increased ENPEP expression were detected in trophoblast differentiation. Knockout of ENPEP in hEPSC-em compromised trophoblast differentiation potency, reduced adhesion and invasion of trophoblastic spheroids, and impeded trophoblastic stem cell (TSC) derivation. Importantly, TET2 was involved in the loss of DNA methylation and activation of ENPEP expression during trophoblast differentiation. TET2-null hEPSC-em failed to produce TSC properly. Collectively, our results illustrated the crucial roles of ENPEP and TET2 in trophoblast fate commitments and the unprecedented TET2-mediated loss of DNA methylation in ENPEP promoter.


Asunto(s)
Diferenciación Celular , Metilación de ADN , Proteínas de Unión al ADN , Dioxigenasas , Proteínas Proto-Oncogénicas , Trofoblastos , Femenino , Humanos , Embarazo , Blastocisto/metabolismo , Blastocisto/citología , Linaje de la Célula/genética , Dioxigenasas/metabolismo , Dioxigenasas/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Trofoblastos/metabolismo , Trofoblastos/citología
2.
Reproduction ; 165(3): 335-346, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36656637

RESUMEN

In brief: Implantation failure can occur even after the transfer of good-quality embryos. This study showed that the migration of human endometrial stromal cells towards embryonic trophoblasts is higher in women with live births in the first in vitro fertilization cycle than those with repeated implantation failure, suggesting that the chemotactic response of stroma cells is associated with successful pregnancy. Abstract: The success rate of in vitro fertilization (IVF) remains limited in some women despite transfers of good-quality embryos in repeated attempts. There is no reliable tool for assessing endometrial receptivity. This study aimed to assess the interaction between decidualized human primary endometrial stromal cells (1°-EnSC) and human embryonic stem cell-derived trophoblastic spheroids (BAP-EB) and to compare the invasion ability of decidualized 1°-EnSC towards BAP-EB between women attaining live birth in the first IVF cycle and those with repeated implantation failure (RIF). The invasion of the decidualized human endometrial cell line (T-HESC) and 1°-EnSC towards BAP-EB was studied. Real-time quantitative PCR and immunocytochemistry were employed to determine the expression of decidualization markers at mRNA and protein levels, respectively. Trophoblast-like BAP-EB-96h, instead of early trophectoderm (TE)-like BAP-EB-48h, facilitated the invasion ability of decidualized T-HESC and decidualized 1°-EnSC. Human chorionic gonadotropin at supra-physiological levels promoted the invasiveness of decidualized 1°-EnSC. The extent of BAP-EB-96h-induced invasion was significantly stronger in decidualized 1°-EnSC from women who had a live birth in the first IVF cycle when compared to those with RIF. While no difference was found in the expression of decidualization markers, PRL and IGFBP1 among two groups of women, significantly lower HLA-B was detected in the non-decidualized and decidualized 1°-EnSC from women with RIF. Collectively, the findings suggested that the invasion of decidualized 1°-EnSC towards trophoblast-like BAP-EB-96h was higher in women who had a live birth in the first IVF cycle than those with RIF.


Asunto(s)
Implantación del Embrión , Trofoblastos , Femenino , Humanos , Embarazo , Línea Celular , Gonadotropina Coriónica , Implantación del Embrión/fisiología , Endometrio/metabolismo , Células del Estroma/metabolismo , Trofoblastos/metabolismo , Insuficiencia del Tratamiento
3.
Int J Mol Sci ; 22(19)2021 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-34639069

RESUMEN

The prevalence of type 2 diabetes (T2D) is rapidly increasing across the globe. Fetal exposure to maternal diabetes was correlated with higher prevalence of impaired glucose tolerance and T2D later in life. Previous studies showed aberrant DNA methylation patterns in pancreas of T2D patients. However, the underlying mechanisms remained largely unknown. We utilized human embryonic stem cells (hESC) as the in vitro model for studying the effects of hyperglycemia on DNA methylome and early pancreatic differentiation. Culture in hyperglycemic conditions disturbed the pancreatic lineage potential of hESC, leading to the downregulation of expression of pancreatic markers PDX1, NKX6-1 and NKX6-2 after in vitro differentiation. Genome-wide DNA methylome profiling revealed over 2000 differentially methylated CpG sites in hESC cultured in hyperglycemic condition when compared with those in control glucose condition. Gene ontology analysis also revealed that the hypermethylated genes were enriched in cell fate commitment. Among them, NKX6-2 was validated and its hypermethylation status was maintained upon differentiation into pancreatic progenitor cells. We also established mouse ESC lines at both physiological glucose level (PG-mESC) and conventional hyperglycemia glucose level (HG-mESC). Concordantly, DNA methylome analysis revealed the enrichment of hypermethylated genes related to cell differentiation in HG-mESC, including Nkx6-1. Our results suggested that hyperglycemia dysregulated the epigenome at early fetal development, possibly leading to impaired pancreatic development.


Asunto(s)
Diferenciación Celular/genética , Metilación de ADN , Células Madre Embrionarias/metabolismo , Hiperglucemia/genética , Hiperglucemia/metabolismo , Páncreas/citología , Páncreas/metabolismo , Línea Celular , Células Cultivadas , Biología Computacional/métodos , Diabetes Mellitus Tipo 2 , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucosa/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo
4.
Differentiation ; 105: 33-44, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30599359

RESUMEN

Gap junctional intercellular communication (GJIC) is important for maintaining the pluripotency of mouse embryonic stem cells (mESC). However, human ESC (hESC) have a high level of connexin (Cx) molecules with unknown function. In this study, we found that the major Cx molecule, Cx43, was highly expressed in undifferentiated hESC. It was down-regulated upon spontaneously differentiation by embryoid body formation and induced differentiation along ectoderm, mesoderm and extraembryonic lineages, but up-regulated along endoderm differentiation. The knockdown of Cx43 and GJIC had no effect on the maintenance of hESC, as demonstrated by no morphological changes and similar expression levels of pluripotent markers (OCT4, NANOG, SSEA-3 and SSEA-4) and early differentiation markers (KRT8 and KRT18). Meanwhile, Cx43 knock down had no effect on endodermal markers (SOX17, FOXA2 and CXCR4) expression when hESC were differentiating into definitive endoderm lineage. On the contrary, it led to lower levels of mesodermal markers (CD56, CD34 and PDGFR-α) when cells were undergoing mesoderm differentiation. When compared to control, Cx43 knockdown led to higher attachment rate, HCG secretion and cell invasion of the hESC derived trophoblastic cells. Cx43 knockdown also resulted in up-regulated expressions of placental hormone (ß-hCG) and implantation related genes (LIFR, CDH5, LEP, PGF, TGFBR2). Our study suggested that Cx43 and GJIC had no effect on the undifferentiated growth of hESC but affected specific lineage differentiation.


Asunto(s)
Diferenciación Celular , Conexina 43/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Comunicación Celular , Línea Celular , Linaje de la Célula , Células Cultivadas , Células Madre Embrionarias Humanas/citología , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Trofoblastos/citología , Trofoblastos/efectos de los fármacos
5.
Hum Reprod ; 30(11): 2614-26, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26350609

RESUMEN

STUDY QUESTION: Can human embryonic stem cell-derived trophoblastic spheroids be used to study the early stages of implantation? SUMMARY ANSWER: We generated a novel human embryonic stem cell-derived trophoblastic spheroid model mimicking human blastocysts in the early stages of implantation. WHAT IS KNOWN ALREADY: Both human embryos and choriocarcinoma cell line derived spheroids can attach onto endometrial cells and are used as models to study the early stages of implantation. However, human embryos are limited and the use of cancer cell lines for spheroid generation remains sub-optimal for research. STUDY DESIGN, SIZE, DURATION: Experimental induced differentiation of human embryonic stem cells into trophoblast and characterization of the trophoblast. PARTICIPANTS/MATERIALS, SETTING, METHODS: Trophoblastic spheroids (BAP-EB) were generated by inducing differentiation of a human embryonic stem cell line, VAL3 cells with bone morphogenic factor-4, A83-01 (a TGF-ß inhibitor), and PD173074 (a FGF receptor-3 inhibitor) after embryoid body formation. The expressions of trophoblastic markers and hCG levels were studied by real-time PCR and immunohistochemistry. BAP-EB attachment and invasion assays were performed on different cell lines and primary endometrial cells. MAIN RESULTS AND THE ROLE OF CHANCE: After 48 h of induced differentiation, the BAP-EB resembled early implanting human embryos in terms of size and morphology. The spheroids derived from embryonic stem cells (VAL3), but not from several other cell lines studied, possessed a blastocoel-like cavity. BAP-EB expressed several markers of trophectoderm of human blastocysts on Day 2 of induced differentiation. In the subsequent days of differentiation, the cells of the spheroids differentiated into trophoblast-like cells expressing trophoblastic markers, though at levels lower than that in the primary trophoblasts or in a choriocarcinoma cell line. On Day 3 of induced differentiation, BAP-EB selectively attached onto endometrial epithelial cells, but not other non-endometrial cell lines or an endometrial cell line that had lost its epithelial character. The attachment rates of BAP-EB was significantly higher on primary endometrial epithelial cells (EEC) taken from 7 days after hCG induction of ovulation (hCG+7 day) when compared with that from hCG+2 day. The spheroids also invaded through Ishikawa cells and the primary endometrial stromal cells in the co-culture. LIMITATIONS, REASONS FOR CAUTION: The attachment rates of BAP-EB were compared between EEC obtained from Day 2 and Day 7 of the gonadotrophin stimulated cycle, but not the natural cycles. WIDER IMPLICATIONS OF THE FINDINGS: BAP-EB have the potential to be used as a test for predicting endometrial receptivity in IVF cycles and provide a novel approach to study early human implantation, trophoblastic cell differentiation and trophoblastic invasion into human endometrial cells.


Asunto(s)
Implantación del Embrión , Células Madre Embrionarias Humanas , Modelos Biológicos , Esferoides Celulares , Trofoblastos , Línea Celular , Humanos
6.
Fertil Steril ; 120(2): 268-276, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36940739

RESUMEN

OBJECTIVE: To evaluate the attachment rate of a human embryonic stem cell-derived trophoblastic spheroid onto endometrial epithelial cells in predicting the cumulative live birth rate of an in vitro fertilization (IVF) cycle. DESIGN: A prospective observational study. SETTING: University hospital and research laboratory. PATIENT(S): A total of 240 infertile women from 2017-2021. INTERVENTION(S): Infertile women with regular cycles attending for IVF were recruited. An endometrial aspirate was collected from a natural cycle 1 month before IVF to determine the BAP-EB attachment rate. MAIN OUTCOME MEASURE(S): Cumulative live birth rates from a stimulated cycle and its derived frozen embryo transfer cycles within 6 months of ovarian stimulation were obtained. RESULT(S): The BAP-EB attachment rate in women who attained a cumulative live birth was similar to that in those who did not. When women were stratified by age into <35 years and ≥35 years, the BAP-EB attachment rate was significantly higher only in women aged ≥35 years having a live birth when compared with those in the same age group without a live birth. Receiver operating characteristic curve analysis of BAP-EB attachment rate in predicting cumulative live birth showed the areas under the curve of 0.559 (95% confidence interval [CI], 0.479-0.639), 0.448 (95% CI, 0.310-0.585), and 0.613 (95% CI, 0.517-0.710) for all ages, an age of <35 years, and an age of ≥35 years, respectively. CONCLUSION(S): The BAP-EB attachment rate offers only a very modest prediction of the cumulative live birth rate in women aged ≥35 years undergoing IVF. CLINICAL TRIAL REGISTRATION NUMBER: NCT02713854 (https://clinicaltrials.gov/ct2/show/NCT02713854; Date of registration, March 21, 2016; date of enrollment of the first subject, August 1, 2017).


Asunto(s)
Infertilidad Femenina , Nacimiento Vivo , Embarazo , Humanos , Femenino , Infertilidad Femenina/diagnóstico , Infertilidad Femenina/terapia , Fertilización In Vitro , Transferencia de Embrión , Tasa de Natalidad , Inducción de la Ovulación , Índice de Embarazo
7.
Adv Sci (Weinh) ; 10(11): e2204797, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36775869

RESUMEN

Human expanded potential stem cells (hEPSC) have been derived from human embryonic stem cells and induced pluripotent stem cells. Here direct derivation of hEPSC from human pre-implantation embryos is reported. Like the reported hEPSC, the embryo-derived hEPSC (hEPSC-em) exhibit a transcriptome similar to morula, comparable differentiation potency, and high genome editing efficiency. Interestingly, the hEPSC-em show a unique H3 lysine-4 trimethylation (H3K4me3) open chromatin conformation; they possess a higher proportion of H3K4me3 bound broad domain (>5 kb) than the reported hEPSC, naive, and primed embryonic stem cells. The open conformation is associated with enhanced trophoblast differentiation potency with increased trophoblast gene expression upon induction of differentiation and success in derivation of trophoblast stem cells with bona fide characteristics. Hippo signaling is specifically enriched in the H3K4me3 broad domains of the hEPSC-. Knockout of the Hippo signaling gene, YAP1 abolishes the ability of the embryo-derived EPSC to form trophoblast stem cells.


Asunto(s)
Cromatina , Trofoblastos , Humanos , Trofoblastos/metabolismo , Diferenciación Celular/genética , Embrión de Mamíferos , Células Madre Embrionarias
8.
Methods Mol Biol ; 2520: 181-187, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35218527

RESUMEN

The use of human embryos for studying the early implantation processes and trophoblast is restricted by ethical concerns. The development of models mimicking the peri-implantation embryos is critical for understanding the physiology of human embryos and many pathophysiological disorders including recurrent implantation failure and miscarriage. Three-dimensional (3D) models of trophoblastic spheroids have been successfully derived from human embryonic stem cells (hESC). Simultaneous activation of the BMP pathway and blockage of the Activin/Nodal pathway favor the differentiation of hESC into trophoblast. Here we describe a 3D trophectoderm differentiation protocol with the use of BAP (BMP4, A83-01, and PD173074) to generate hESC-derived trophectoderm spheroids (BAP-EB). BAP-EB is highly reproducible and exhibits morphological and transcriptomic similarities to human early blastocysts.


Asunto(s)
Células Madre Embrionarias Humanas , Blastocisto , Diferenciación Celular , Implantación del Embrión , Humanos , Trofoblastos
9.
Genes (Basel) ; 12(10)2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34680943

RESUMEN

Pluripotent stem cells (PSCs) hold great promise in cell-based therapy because of their pluripotent property and the ability to proliferate indefinitely. Embryonic stem cells (ESCs) derived from inner cell mass (ICM) possess unique cell cycle control with shortened G1 phase. In addition, ESCs have high expression of homologous recombination (HR)-related proteins, which repair double-strand breaks (DSBs) through HR or the non-homologous end joining (NHEJ) pathway. On the other hand, the generation of induced pluripotent stem cells (iPSCs) by forced expression of transcription factors (Oct4, Sox2, Klf4, c-Myc) is accompanied by oxidative stress and DNA damage. The DNA repair mechanism of DSBs is therefore critical in determining the genomic stability and efficiency of iPSCs generation. Maintaining genomic stability in PSCs plays a pivotal role in the proliferation and pluripotency of PSCs. In terms of therapeutic application, genomic stability is the key to reducing the risks of cancer development due to abnormal cell replication. Over the years, we and other groups have identified important regulators of DNA damage response in PSCs, including FOXM1, SIRT1 and PUMA. They function through transcription regulation of downstream targets (P53, CDK1) that are involved in cell cycle regulations. Here, we review the fundamental links between the PSC-specific HR process and DNA damage response, with a focus on the roles of FOXM1 and SIRT1 on maintaining genomic integrity.


Asunto(s)
Ciclo Celular , Daño del ADN , Células Madre Pluripotentes/metabolismo , Animales , Humanos , Células Madre Pluripotentes/fisiología
10.
Aging (Albany NY) ; 12(8): 7431-7447, 2020 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-32335545

RESUMEN

Sirt1 facilitates the reprogramming of mouse somatic cells into induced pluripotent stem cells (iPSCs). It is regulated by micro-RNA and reported to be a target of miR-135a. However, their relationship and roles on cellular reprogramming remain unknown. In this study, we found negative correlations between miR-135a and Sirt1 during mouse embryonic stem cells differentiation and mouse embryonic fibroblasts reprogramming. We further found that the reprogramming efficiency was reduced by the overexpression of miR-135a precursor but induced by the miR-135a inhibitor. Co-immunoprecipitation followed by mass spectrometry identified 21 SIRT1 interacting proteins including KU70 and WRN, which were highly enriched for DNA damage repair. In accordance, Sirt1 activator resveratrol reduced DNA damage during the reprogramming process. Wrn was regulated by miR-135a and resveratrol partly rescued the impaired reprogramming efficiency induced by Wrn knockdown. This study showed Sirt1, being partly regulated by miR-135a, bound proteins involved in DNA damage repair and enhanced the iPSCs production.


Asunto(s)
ADN/genética , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/genética , Sirtuina 1/genética , Animales , Diferenciación Celular , Reprogramación Celular/genética , Daño del ADN , Células Madre Pluripotentes Inducidas/citología , Ratones , MicroARNs/biosíntesis , Modelos Animales , Sirtuina 1/biosíntesis
11.
Fertil Steril ; 114(3): 653-664.e6, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32444068

RESUMEN

OBJECTIVE: To study the use of human embryonic stem cell-derived trophoblastic spheroids (BAP-EB) as human blastocyst surrogates for studying early implantation and trophoblast development. DESIGN: Laboratory study. SETTING: University research laboratory. PATIENT(S): Infertile in vitro fertilization patients donating endometrial aspirates and human embryonic stem cells (hESCs: VAL3 and H9/WA09). INTERVENTION(S): In BAP-EB derived from hESC, transcriptomes analyzed by next-generation RNA sequencing, effects of Hippo signaling pathway studied by a YAP inhibitor, comparison of attachment of BAP-EB onto primary endometrial epithelial cells (EEC) collected at prereceptive and receptive phases, and antibody blocking assay used to study the molecule(s) involved in BAP-EB attachment. MAIN OUTCOME MEASURE(S): Gene expression profiles and endometrial cell attachment rates. RESULT(S): The BAP-EB differentiation protocol for VAL3 could be used to induce trophoblast differentiation in another hESC line, H9. Transcriptomic analysis showed that the epiblast signature gene expression was reduced while that of the trophoblast was induced during BAP-EB differentiation. Specifically, trophectoderm signature genes were induced in BAP-EB at 48 hours and 72 hours after induction of differentiation. The Hippo signaling pathway was one of the pathways induced during BAP-EB differentiation, and YAP1 inhibitor statistically significantly reduced attachment, outgrowth, and trophoblast gene expressions of BAP-EB. A statistically significantly higher number of BAP-EB derived from both VAL3 and H9 attached onto receptive EEC than prereceptive EEC. The antibody blocking assay demonstrated that endometrial E-cadherin might be critical in early implantation. CONCLUSION(S): The data suggest that BAP-EB possesses a trophectoderm-like signature, which supports the use of BAP-EB as a blastocyst surrogate for the study of trophoblast development and endometrial receptivity.


Asunto(s)
Blastocisto/fisiología , Implantación del Embrión , Células Madre Embrionarias/fisiología , Endometrio/fisiología , Blastocisto/citología , Adhesión Celular , Diferenciación Celular , Línea Celular , Técnicas de Cocultivo , Endometrio/citología , Células Endoteliales/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Transducción de Señal , Esferoides Celulares , Factores de Tiempo , Transcriptoma , Trofoblastos/fisiología
12.
Environ Int ; 130: 104885, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31195220

RESUMEN

Animal and epidemiological studies demonstrated association of persistent exposure of TCDD, an endocrine disrupting chemical, to susceptibility of type 2 diabetes (T2D). High doses of TCDD were commonly employed in experimental animals to illustrate its diabetogenic effects. Data linking the epigenetic effects of low doses of TCDD on embryonic cells to T2D susceptibility risks is very limited. To address whether low dose exposure to TCDD would affect pancreatic development, hESCs pretreated with TCDD at concentrations similar to human exposure were differentiated towards pancreatic lineage cells, and their global DNA methylation patterns were determined. Our results showed that TCDD-treated hESCs had impaired pancreatic lineage differentiation potentials and altered global DNA methylation patterns. Four of the hypermethylated genes (PRKAG1, CAPN10, HNF-1B and MAFA) were validated by DNA bisulfite sequencing. PRKAG1, a regulator in the AMPK signaling pathway critical for insulin secretion, was selected for further functional study in the rat insulinoma cell line, INS-1E cells. TCDD treatment induced PRKAG1 hypermethylation in hESCs, and the hypermethylation was maintained after pancreatic progenitor cells differentiation. Transient Prkag1 knockdown in the INS-1E cells elevated glucose stimulated insulin secretions (GSIS), possibly through mTOR signaling pathway. The current study suggested that early embryonic exposure to TCDD might alter pancreatogenesis, increasing the risk of T2D.


Asunto(s)
Células Madre Embrionarias/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Células Secretoras de Insulina/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Metilación de ADN/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/citología , Ratas
13.
PLoS One ; 7(9): e45633, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029150

RESUMEN

Forced-expression of transcription factors can reprogram somatic cells into induced pluripotent stem cells (iPSC). Recent studies show that the reprogramming efficiency can be improved by inclusion of small molecules that regulate chromatin modifying enzymes. We report here that sirtuin 1 (SIRT1), a member of the sirtuin family of NAD(+)-dependent protein deacetylases, is involved in iPSC formation. By using an efficient mouse secondary fibroblast reprogramming system with doxycycline (DOX) inducible Yamanaka's transcription factors delivered by piggyBac (PB) transposition (2°F/1B MEF), we show that SIRT1 knockdown decreased while resveratrol (RSV) increased the efficiency of iPSC formation. The treatments were associated with altered acetylated p53 and its downstream Nanog but not p21 expression. The stimulatory effect was also confirmed by SIRT1 over-expression, which stimulated the formation of colonies with induced Nanog and reduced p21 expression. Furthermore, the effects of RSV and SIRT1 knockdown on reprogramming were most pronounced during the initiation phase of reprogramming. MicroRNA-34a is a known regulator of SIRT1. Its inhibitor increased, while its mimics reduced iPSC formation. The stimulatory effect of SIRT1 during reprogramming was also confirmed in the primary MEF. RSV increased while tenovin-6, a small molecule that activates p53 through SIRT1 inhibition, suppressed reprogramming. In conclusion, SIRT1 enhances iPSC generation, in part, through deacetylation of p53, inhibition of p21 and enhancement of Nanog expression.


Asunto(s)
Diferenciación Celular/fisiología , Embrión de Mamíferos/citología , MicroARNs/metabolismo , Células Madre Pluripotentes/citología , Sirtuina 1/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Western Blotting , Células Cultivadas , Regulación hacia Abajo , Fibroblastos/citología , Humanos , Ratones , Reacción en Cadena de la Polimerasa , Regulación hacia Arriba
14.
PLoS One ; 6(6): e21602, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21738728

RESUMEN

Nucleosome assembly proteins play important roles in chromatin remodeling, which determines gene expression, cell proliferation and terminal differentiation. Testis specific protein, Y-encoded-like 2 (TSPYL2) is a nucleosome assembly protein expressed in neuronal precursors and mature neurons. Previous studies have shown that TSPYL2 binds cyclin B and inhibits cell proliferation in cultured cells suggesting a role in cell cycle regulation. To investigate the physiological significance of TSPYL2 in the control of cell cycle, we generated mice with targeted disruption of Tspyl2. These mutant mice appear grossly normal, have normal life span and do not exhibit increased tumor incidence. To define the role of TSPYL2 in DNA repair, checkpoint arrest and apoptosis, primary embryonic fibroblasts and thymocytes from Tspyl2 deficient mice were isolated and examined under unperturbed and stressed conditions. We show that mutant fibroblasts are impaired in G1 arrest under the situation of DNA damage induced by gamma irradiation. This is mainly attributed to the defective activation of p21 transcription despite proper p53 protein accumulation, suggesting that TSPYL2 is additionally required for p21 induction. TSPYL2 serves a biological role in maintaining the G1 checkpoint under stress condition.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Northern Blotting , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Células Cultivadas , Daño del ADN/genética , Daño del ADN/fisiología , Citometría de Flujo , Fase G1/genética , Fase G1/fisiología , Inmunohistoquímica , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA