Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
1.
Biochem Biophys Res Commun ; 613: 187-192, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35598374

RESUMO

Histone lysine crotonylation (Kcr) is a novel hydrophobic histone acylation modification, and we recently report its crucial roles in neural differentiation. However, it is still unclear how histone Kcr involve in early neural commitment. Here, we systematically investigate the H3K9cr landscapes during neuroectodermal differentiation of pluripotent P19 embryonal carcinoma cells (ECCs). We reveal that the genome-wide changes in H3K9cr favor neural fate specification, and identify potential co-factors binding H3K9cr. We also uncover that H3K9cr collaborates with H3K9ac to regulate gene expression changes. Our results provide novel insights into the epigenetic mechanisms underlying neural commitment.


Assuntos
Histonas , Lisina , Diferenciação Celular , Células-Tronco de Carcinoma Embrionário/metabolismo , Epigênese Genética , Histonas/metabolismo , Lisina/metabolismo , Processamento de Proteína Pós-Traducional
2.
Toxicology ; 470: 153138, 2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-35219798

RESUMO

Bisphenol A (2,2-bis(4'-hydroxyphenyl) propane, BPA) is a well-known endocrine-disrupting compound that is widely used in various daily products and exhibits embryonic development toxicity and genotoxicity. However, the affected signaling pathways involved in embryonic development especially the interactions of involved proteins remain unclear. In our previous study (Ge et al., 2021), BPA induces DNA damage and apoptosis in Xenopus embryos, resulting in multiple malformations of larvae. However, the signaling pathways induced for apoptosis response to DNA damage are still not well elucidated. Here, we systematically elucidated the enriched pathways affected by BPA and illustrated the interactions of involved proteins. Results indicated that BPA affected multiple embryonic development pathways including Hippo, TGF-ß, Wnt, and Notch pathways. Furthermore, the protein-protein interaction network suggested that the c-Abl/YAPY357/p73 pathway may play a key role in apoptosis induction in response to DNA damage. P19 embryonal carcinoma stem cells, as a developmental toxicity model, were treated with different BPA concentrations to establish an in vitro model to verify the role of the c-Abl/YAPY357/p73 pathway in apoptosis. BPA triggered DNA damage and significantly upregulated the expression levels of c-Abl, phosphorylated YAPY357, phosphorylated p73Y99, and cleaved caspase-3 protein (p < 0.05), thus decreasing cell viability and transcriptionally activating the p73 target genes Bax and Puma. These data suggested that BPA activated the c-Abl/YAPY357/p73 pathway in response to DNA damage. Imatinib, an inhibitor of tyrosine kinase c-Abl, significantly downregulated the elevated expression levels of p-YAPY357, p-p73Y99 and cleaved caspase-3 (p < 0.05) caused by BPA and then ameliorated the cell index of P19 cells in the BPA-treated group. Therefore, this substance restrained the phosphokinase activity of c-Abl and suppressed the c-Abl/YAPY357/p73 pathway. Results showed that the c-Abl/YAPY357/p73 pathway served as a mechanism for caspase-3 activation that induced the apoptosis response to DNA damage stress.


Assuntos
Proteínas de Ligação a DNA , Proteínas Nucleares , Apoptose/genética , Compostos Benzidrílicos , Caspase 3/genética , Dano ao DNA , Proteínas de Ligação a DNA/genética , Células-Tronco de Carcinoma Embrionário/metabolismo , Proteínas Nucleares/genética , Fenóis , Proteína Tumoral p73/genética , Proteínas Supressoras de Tumor/metabolismo
3.
Cells ; 10(11)2021 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-34831070

RESUMO

Embryonic cancer stem cells (CSCs) can differentiate into any cancer type. Targeting CSC using natural compounds is a good approach as it suppresses cancer recurrence with fewer adverse effects, and methylsulfonylmethane (MSM) is a sulfur-containing compound with well-known anticancer activities. This study determined the mechanistic aspects of the anticancer activity of MSM. We used Western blotting and real-time qPCR for molecular signaling studies and conducted flow cytometry for analyzing the processes in cells. Our results suggested an inhibition in the expression of CSC markers and Wnt/ß-catenin signaling. MSM induced TRAIL-mediated extrinsic apoptosis in NCCIT and NTERA-2 cells rather than an intrinsic pathway. Inhibition of iron metabolism-dependent reactive oxygen species (ROS) generation takes part in TRAIL-mediated apoptosis induction by MSM. Suppressing iron metabolism by MSM also regulated p38/p53/ERK signaling and microRNA expressions, such as upregulating miR-130a and downregulating miR-221 and miR-222, which resulted in TRAIL induction and thereby extrinsic pathway of apoptosis. Hence, MSM could be a good candidate for neoadjuvant therapy by targeting CSCs by inhibiting iron metabolism.


Assuntos
Apoptose , Dimetil Sulfóxido/farmacologia , Células-Tronco de Carcinoma Embrionário/patologia , Ferro/metabolismo , Sulfonas/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Dano ao DNA , Células-Tronco de Carcinoma Embrionário/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Modelos Biológicos , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Sci Rep ; 11(1): 20075, 2021 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-34625606

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and behavioral changes. Extracellular deposition of amyloid plaques (Aß) and intracellular deposition of neurofibrillary tangles in neurons are the major pathogenicities of AD. However, drugs targeting these therapeutic targets are not effective. Therefore, novel targets for the treatment of AD urgently need to be identified. Expression of the mesoderm-specific transcript (Mest) is regulated by genomic imprinting, where only the paternal allele is active for transcription. We identified hypermethylation on the Mest promoter, which led to a reduction in Mest mRNA levels and activation of Wnt signaling in brain tissues of AD patients. Mest knockout (KO) using the CRIPSR/Cas9 system in mouse embryonic stem cells and P19 embryonic carcinoma cells leads to neuronal differentiation arrest. Depletion of Mest in primary hippocampal neurons via lentivirus expressing shMest or inducible KO system causes neurodegeneration. Notably, depletion of Mest in primary cortical neurons of rats leads to tau phosphorylation at the S199 and T231 sites. Overall, our data suggest that hypermethylation of the Mest promoter may cause or facilitate the progression of AD.


Assuntos
Doença de Alzheimer/patologia , Metilação de DNA , Células-Tronco Embrionárias/patologia , Neurônios/patologia , Regiões Promotoras Genéticas , Proteínas/genética , Via de Sinalização Wnt , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Células-Tronco de Carcinoma Embrionário/metabolismo , Células-Tronco de Carcinoma Embrionário/patologia , Células-Tronco Embrionárias/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Camundongos , Neurônios/metabolismo , Fosforilação , Proteínas/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
5.
Development ; 148(9)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33912935

RESUMO

In response to signals from the embryonic testis, the germ cell intrinsic factor NANOS2 coordinates a transcriptional program necessary for the differentiation of pluripotent-like primordial germ cells toward a unipotent spermatogonial stem cell fate. Emerging evidence indicates that genetic risk factors contribute to testicular germ cell tumor initiation by disrupting sex-specific differentiation. Here, using the 129.MOLF-Chr19 mouse model of testicular teratomas and a NANOS2 reporter allele, we report that the developmental phenotypes required for tumorigenesis, including failure to enter mitotic arrest, retention of pluripotency and delayed sex-specific differentiation, were exclusive to a subpopulation of germ cells failing to express NANOS2. Single-cell RNA sequencing revealed that embryonic day 15.5 NANOS2-deficient germ cells and embryonal carcinoma cells developed a transcriptional profile enriched for MYC signaling, NODAL signaling and primed pluripotency. Moreover, lineage-tracing experiments demonstrated that embryonal carcinoma cells arose exclusively from germ cells failing to express NANOS2. Our results indicate that NANOS2 is the nexus through which several genetic risk factors influence tumor susceptibility. We propose that, in the absence of sex specification, signals native to the developing testis drive germ cell transformation.


Assuntos
Diferenciação Celular , Neoplasias Embrionárias de Células Germinativas , Diferenciação Sexual , Neoplasias Testiculares , Animais , Diferenciação Celular/genética , Proliferação de Células , Células-Tronco de Carcinoma Embrionário/metabolismo , Células Germinativas Embrionárias , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Proteínas de Ligação a RNA , Transdução de Sinais , Espermatogônias/metabolismo , Teratoma
6.
Int J Mol Sci ; 22(5)2021 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-33668324

RESUMO

FOXC1, a transcription factor involved in cell differentiation and embryogenesis, is demonstrated to be a negative regulator of Nanog in this study. FOXC1 is up-regulated in retinoic acid-induced differentiation of F9 Embryonal Carcinoma (EC) cells; furthermore, FOXC1 specifically inhibits the core pluripotency factor Nanog by binding to the proximal promoter. Overexpression of FOXC1 in F9 or knockdown in 3T3 results in the down-regulation or up-regulation of Nanog mRNA and proteins, respectively. In order to explain the mechanism by which FOXC1 inhibits Nanog expression, we identified the co-repressor HDAC2 from the FOXC1 interactome. FOXC1 recruits HDAC2 to Nanog promoter to decrease H3K27ac enrichment, resulting in transcription inhibition of Nanog. To the best of our knowledge, this is the first report that FOXC1 is involved in the epigenetic regulation of gene expression.


Assuntos
Células-Tronco de Carcinoma Embrionário/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histona Desacetilase 2/metabolismo , Proteína Homeobox Nanog/genética , Regiões Promotoras Genéticas , Tretinoína/farmacologia , Animais , Antineoplásicos/farmacologia , Células-Tronco de Carcinoma Embrionário/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário/patologia , Epigênese Genética , Fatores de Transcrição Forkhead/genética , Células HEK293 , Histona Desacetilase 2/genética , Humanos , Camundongos , Células NIH 3T3 , Proteína Homeobox Nanog/metabolismo
7.
Mol Med Rep ; 22(6): 4675-4684, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33173954

RESUMO

As a single cardiac malformation, ventricular septal defect (VSD) is the most common form of congenital heart disease. However, the precise molecular mechanisms underlying VSD are not completely understood. Numerous microRNAs (miRs/miRNAs) are associated with ventricular septal defects. miR-29c inhibits the proliferation and promotes the apoptosis and differentiation of P19 embryonal carcinoma cells, possibly via suppressing Wnt4 signaling. However, to the best of our knowledge, no in vivo studies have been published to determine whether overexpression of miR-29c leads to developmental abnormalities. The present study was designed to observe the effect of miRNA-29c on cardiac development and its possible mechanism in vivo. Zebrafish embryos were microinjected with different doses (1, 1.6 and 2 µmol) miR-29c mimics or negative controls, and hatchability, mortality and cardiac malformation were subsequently observed. The results showed that in zebrafish embryos, miR-29c overexpression attenuated heart development in a dose-dependent manner, manifested by heart rate slowdown, pericardial edema and heart looping disorder. Further experiments showed that overexpression of miR-29c was associated with the Wnt4/β-catenin signaling pathway to regulate zebrafish embryonic heart development. In conclusion, the present results demonstrated that miR-29c regulated the lateral development and cardiac circulation of zebrafish embryo by targeting Wnt4.


Assuntos
Comunicação Interventricular/metabolismo , MicroRNAs/metabolismo , Proteína Wnt4/metabolismo , Animais , Apoptose/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Células-Tronco de Carcinoma Embrionário/metabolismo , Coração/embriologia , Comunicação Interventricular/genética , MicroRNAs/genética , Transdução de Sinais/genética , Proteína Wnt4/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
8.
Andrology ; 8(6): 1844-1858, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32741077

RESUMO

BACKGROUND: Testicular germ cell tumors (TGCTs) are the most common malignant cancer in young men. Although TGCTs are generally responsive to platinum-based chemotherapy particularly cisplatin, acquired resistance in patients with metastasis still occurs resulting in poor prognosis. Specifically, differentiation of embryonal carcinoma (EC) cells, the stem cells of TGCTs, can lead to the reduction of cisplatin responsiveness. Therefore, novel therapeutic strategies for TGCTs are needed. System L amino acid transporters have been reported to be up-regulated and to play an important role in tumorigenesis. However, expression and role of system L amino acid transporters in TGCTs remain elusive. MATERIALS AND METHODS: Expression of system L amino acid transporters was analyzed in TGCT samples from The Cancer Genome Atlas (TCGA). Expression of LAT1, LAT2, and 4F2hc was examined in human embryonal carcinoma cell line NTERA2. Roles of system L amino acid transporters on NTERA2 cell survival, cell proliferation, pluripotency, and cisplatin sensitivity were evaluated. RESULTS: Based upon TCGA datasets, we found that two isoforms of system L (LAT1 and LAT2) and their chaperone protein 4F2hc are highly expressed in EC samples compared with other groups. Treatment with the system L inhibitor BCH significantly suppressed leucine uptake into the pluripotent EC cell line NTERA2. The malignant phenotypes including cell viability, cell proliferation, and clonal ability were decreased following BCH treatment. Nonetheless, system L inhibition did not alter expression of stemness genes in NTERA2 cells. After NTERA2 differentiation, expressions of LAT1 and LAT2 were decreased. Finally, co-administration of BCH enhanced cisplatin sensitivity in both undifferentiated and differentiated cells. These effects were associated with the reduction in p70S6K phosphorylation. CONCLUSION: Taken together, these results shed light on the roles of system L amino acid transporters in TGCTs. Therefore, system L amino acid transporters could provide novel therapeutic targets for treatment against TGCTs.


Assuntos
Sistema L de Transporte de Aminoácidos/biossíntese , Sistema L de Transporte de Aminoácidos/metabolismo , Carcinoma Embrionário/patologia , Células-Tronco de Carcinoma Embrionário/metabolismo , Neoplasias Testiculares/patologia , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Antineoplásicos/farmacologia , Carcinogênese/patologia , Carcinoma Embrionário/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Cadeia Pesada da Proteína-1 Reguladora de Fusão/biossíntese , Humanos , Transportador 1 de Aminoácidos Neutros Grandes/biossíntese , Masculino , Neoplasias Testiculares/tratamento farmacológico
9.
RNA Biol ; 17(11): 1613-1624, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32372724

RESUMO

PIWI homologs constitute a subclass of the Argonaute family. Traditionally, they have been shown to associate with a specific class of small RNAs, piRNAs, to suppress transposable elements and protect genomic integrity in germ cells. Recent studies imply that PIWI proteins may also exert important biological functions in somatic contexts, including the brain. However, their exact role in neural development remains unknown. Hence we investigated whether PIWI proteins are involved in neuronal differentiation. By using an established cell model for studying neurogenesis, NTera2/D1 (NT2) cells, we found that a particular PIWI homolog, PIWIL4 was increasingly upregulated throughout the course of all-trans retinoic acid (RA)-mediated neuronal differentiation. During this process, PIWIL4 knockdown led to partial recovery of embryonic stem cell markers, while suppressing RA-induced expression of neuronal markers. Consistently, PIWIL4 overexpression further elevated their expression levels. Furthermore, co-immunoprecipitation revealed an RA-induced interaction between PIWIL4 and the H3K27me3 demethylase UTX. Chromatin immunoprecipitation showed that this interaction could be essential for the removal of H3K27me3 from the promoters of RA-inducible genes. By a similar mechanism, PIWIL4 knockdown also suppressed the expression of PTN and NLGN3, two important neuronal factors secreted to regulate glioma activity. We further noted that the conditioned medium collected from PIWIL4-silenced NT2 cells significantly reduced the proliferation of glioma cells. Thus, our data suggest a novel somatic role of PIWIL4 in modulating the expression of neuronal genes that can be further characterized to promote neuronal differentiation and to modulate the activity of glioma cells.


Assuntos
Diferenciação Celular/genética , Células-Tronco de Carcinoma Embrionário/metabolismo , Células-Tronco de Carcinoma Embrionário/patologia , Neurônios/metabolismo , Proteínas de Ligação a RNA/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Histona Desmetilases/metabolismo , Histonas/metabolismo , Humanos , Neurônios/citologia , Ligação Proteica , Proteínas de Ligação a RNA/metabolismo , Transcriptoma
10.
J Mol Biol ; 432(7): 2271-2288, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32105733

RESUMO

R-loops are a prevalent class of non-B DNA structures that have been associated with both positive and negative cellular outcomes. DNA:RNA immunoprecipitation (DRIP) approaches based on the anti-DNA:RNA hybrid S9.6 antibody revealed that R-loops form dynamically over conserved genic hotspots. We have developed an orthogonal approach that queries R-loops via the presence of long stretches of single-stranded DNA on their looped-out strand. Nondenaturing sodium bisulfite treatment catalyzes the conversion of unpaired cytosines to uracils, creating permanent genetic tags for the position of an R-loop. Long-read, single-molecule PacBio sequencing allows the identification of R-loop 'footprints' at near nucleotide resolution in a strand-specific manner on long single DNA molecules and at ultra-deep coverage. Single-molecule R-loop footprinting coupled with PacBio sequencing (SMRF-seq) revealed a strong agreement between S9.6-based and bisulfite-based R-loop mapping and confirmed that R-loops form over genic hotspots, including gene bodies and terminal gene regions. Based on the largest single-molecule R-loop dataset to date, we show that individual R-loops form nonrandomly, defining discrete sets of overlapping molecular clusters that pileup through larger R-loop zones. R-loops most often map to intronic regions and their individual start and stop positions do not match with intron-exon boundaries, reinforcing the model that they form cotranscriptionally from unspliced transcripts. SMRF-seq further established that R-loop distribution patterns are not simply driven by intrinsic DNA sequence features but most likely also reflect DNA topological constraints. Overall, DRIP-based and SMRF-based approaches independently provide a complementary and congruent view of R-loop distribution, consolidating our understanding of the principles underlying R-loop formation.


Assuntos
DNA/química , Células-Tronco de Carcinoma Embrionário/metabolismo , Estruturas R-Loop , RNA/química , Análise de Célula Única/métodos , Transcrição Gênica , Células-Tronco de Carcinoma Embrionário/citologia , Humanos
11.
Sci Rep ; 9(1): 11928, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31417131

RESUMO

Human embryonal carcinoma (EC) cells comprise the pluripotent stem cells of malignant non-seminomatous germ cell tumors (GCTs) and represent the malignant counterpart of embryonic stem cells (ESCs). WNT/ß-catenin signaling has been implicated in regulating adult and embryonic stem cells although its role in EC cells is less investigated. Here, we studied WNT signaling in a panel of representative pluripotent and nullipotent human EC cell lines. We found that EC cell lines show distinct levels of intrinsic WNT signaling and respond differently to ectopic WNT activation. Short-term activation of WNT signaling induced a differentiation-response in the pluripotent EC cells (NT2 and NCCIT) whereas the nullipotent EC cells (TERA1 and 2102Ep) were refractory and maintained high levels of OCT4 and SSEA4 expression. Long-term activation of WNT signaling in NCCIT and, to a lesser extent, TERA1 cells led to (re)gain of OCT4 expression and a switch from SSEA4 to SSEA1 surface antigens ultimately resulting in OCT4+/SSEA4-/SSEA1+ profile. Cisplatin treatment indicated that the OCT4+/SSEA4-/SSEA1+ NCCIT cells became more resistant to chemotherapy treatment. Our findings are of particular interest for the GCT and ES cell biology and shed light on the role of WNT signaling in human EC cells.


Assuntos
Técnicas de Cultura de Células , Células-Tronco de Carcinoma Embrionário/metabolismo , Células-Tronco de Carcinoma Embrionário/patologia , Via de Sinalização Wnt , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Antígenos Embrionários Estágio-Específicos/metabolismo , Fatores de Tempo , Via de Sinalização Wnt/efeitos dos fármacos
12.
J Vis Exp ; (146)2019 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-31081818

RESUMO

The P19 cell line derived from a mouse embryo-derived teratocarcinoma has the ability to differentiate into the three germ layers. In the presence of retinoic acid (RA), the suspension cultured P19 cell line is induced to differentiate into neurons. This phenomenon is extensively investigated as a neurogenesis model in vitro. Therefore, the P19 cell line is very useful for molecular and cellular studies associated with neurogenesis. However, protocols for neuronal differentiation of P19 cell line described in the literature are very complex. The method developed in this study are simple and will play a part in elucidating the molecular mechanisms in neurodevelopmental abnormalities and neurodegenerative diseases.


Assuntos
Células-Tronco de Carcinoma Embrionário/patologia , Neurogênese , Animais , Diferenciação Celular/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário/metabolismo , Processamento de Imagem Assistida por Computador , Camundongos , Neurogênese/efeitos dos fármacos , Tretinoína/farmacologia
13.
Biochem Biophys Res Commun ; 512(3): 629-634, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30914194

RESUMO

During brain development, the expression of promyelocytic leukemia zinc finger (Plzf) in neural stem cells is precisely controlled to maintain the balance between neural stem cell self-renewal and differentiation. However, the mechanism underlying transcriptional regulation of Plzf in neural stem cell is still unclear. Herein, using P19 embryonal carcinoma cells as a model, we observed that Plzf expression was induced in the P19-derived embryonic bodies, which enrich neural stem-like cell populations, as demonstrated by the expression of neural stem cell markers, Nestin and Sox2. We then characterized the Plzf promoter and identified two E2f1 binding sites (-755/-751 and -53/-49, the transcription start site was designated as +1) are important for the activation of Plzf promoter. Finally, we found that the induction of Plzf in the neural stem-like cells derived from pluripotent P19 cells is decrease by E2f1 knockdown. Taken together, we conclude that E2f1 is an important transcription factor that regulates Plzf transcription and may involve in maintaining the self-renewal ability of neural stem cells.


Assuntos
Fator de Transcrição E2F1/metabolismo , Células-Tronco de Carcinoma Embrionário/patologia , Regulação Neoplásica da Expressão Gênica , Células-Tronco Neurais/patologia , Proteína com Dedos de Zinco da Leucemia Promielocítica/genética , Animais , Linhagem Celular Tumoral , Células-Tronco de Carcinoma Embrionário/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo , Neurogênese , Regiões Promotoras Genéticas , Dedos de Zinco
14.
Neurotoxicol Teratol ; 72: 58-70, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30776472

RESUMO

Lead (Pb) is a teratogen that poses health risks after acute and chronic exposure. Lead is deposited in the bones of adults and is continuously leached into the blood for decades. While this chronic lead exposure can have detrimental effects on adults such as high blood pressure and kidney damage, developing fetuses and young children are particularly vulnerable. During pregnancy, bone-deposited lead is released into the blood at increased rates and can cross the placental barrier, exposing the embryo to the toxin. Embryos exposed to lead display serious developmental and cognitive defects throughout life. Although studies have investigated lead's effect on late-stage embryos, few studies have examined how lead affects stem cell determination and differentiation. For example, it is unknown whether lead is more detrimental to neuronal determination or differentiation of stem cells. We sought to determine the effect of lead on the determination and differentiation of pluripotent embryonic testicular carcinoma (P19) cells into neurons. Our data indicate that lead exposure significantly inhibits the determination of P19 cells to the neuronal lineage by alteration of N-cadherin and Sox2 expression. We also observed that lead significantly alters subsequent neuronal and glial differentiation. Consequently, this research emphasizes the need to reduce public exposure to lead.


Assuntos
Caderinas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Chumbo/toxicidade , Neurônios/efeitos dos fármacos , Fatores de Transcrição SOXB1/metabolismo , Teratogênicos/toxicidade , Animais , Caderinas/genética , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Camundongos , Neurônios/metabolismo , Fatores de Transcrição SOXB1/genética
15.
J Biol Chem ; 294(15): 5914-5922, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30808711

RESUMO

Embryonic stem cells can self-renew and differentiate, holding great promise for regenerative medicine. They also employ multiple mechanisms to preserve the integrity of their genomes. Nucleotide excision repair, a versatile repair mechanism, removes bulky DNA adducts from the genome. However, the dynamics of the capacity of nucleotide excision repair during stem cell differentiation remain unclear. Here, using immunoslot blot assay, we measured repair rates of UV-induced DNA damage during differentiation of human embryonic carcinoma (NTERA-2) cells into neurons and muscle cells. Our results revealed that the capacity of nucleotide excision repair increases as cell differentiation progresses. We also found that inhibition of the apoptotic signaling pathway has no effect on nucleotide excision repair capacity. Furthermore, RNA-Seq-based transcriptomic analysis indicated that expression levels of four core repair factors, xeroderma pigmentosum (XP) complementation group A (XPA), XPC, XPG, and XPF-ERCC1, are progressively up-regulated during differentiation, but not those of replication protein A (RPA) and transcription factor IIH (TFIIH). Together, our findings reveal that increase of nucleotide excision repair capacity accompanies cell differentiation, supported by the up-regulated transcription of genes encoding DNA repair enzymes during differentiation of two distinct cell lineages.


Assuntos
Diferenciação Celular , Reparo do DNA , Células-Tronco de Carcinoma Embrionário/metabolismo , Células Musculares/metabolismo , Proteínas de Neoplasias/metabolismo , Neurônios/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células-Tronco de Carcinoma Embrionário/patologia , Endonucleases/genética , Endonucleases/metabolismo , Humanos , Células Musculares/patologia , Proteínas de Neoplasias/genética , Neurônios/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína de Replicação A/genética , Proteína de Replicação A/metabolismo , Fator de Transcrição TFIIH/genética , Fator de Transcrição TFIIH/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína de Xeroderma Pigmentoso Grupo A/genética , Proteína de Xeroderma Pigmentoso Grupo A/metabolismo
16.
Biochem Biophys Res Commun ; 509(2): 603-610, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30606481

RESUMO

MicroRNA-135a-5p has been reported to play a potential role in the generation of new neurons. However, the underlying targets of miR-135a-5p in regulating neuronal differentiation have been poorly understood. Our study recently has uncovered that Sox6 and CD44 genes were significantly downregulated during neuronal differentiation of P19 cells, a multipotent cell type. We then found that Sox6 directly bound to the promoter of CD44. Importantly, we identified Sox6 as a direct target of miR-135a-5p. Additionally, we demonstrated that miR-135a-5p is crucial for the neuronal differentiation of P19 cells. More significantly, we found that Sox6 overexpression could overturn miR-135a-5p-mediated neuronal differentiation and dendrite development. In conclusion, these findings indicated that miR-135a-5p/Sox6/CD44 axis provides an important molecular target mechanism for neurodifferentiation.


Assuntos
Células-Tronco de Carcinoma Embrionário/patologia , Receptores de Hialuronatos/genética , MicroRNAs/genética , Neurogênese , Fatores de Transcrição SOXD/genética , Animais , Linhagem Celular Tumoral , Células-Tronco de Carcinoma Embrionário/citologia , Células-Tronco de Carcinoma Embrionário/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Receptores de Hialuronatos/metabolismo , Camundongos , Fatores de Transcrição SOXD/metabolismo , Transdução de Sinais
17.
Chemosphere ; 216: 372-378, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30384306

RESUMO

Ambient fine particulate matter (PM2.5) has been found to be associated with congenital heart defects, but the molecular mechanisms remain to be elucidated. Our previous study revealed that extractable organic matter (EOM) from PM2.5 exerted cardiac developmental toxicity in zebrafish embryos. The aim of the current study is to explore the effects of EOM on cardiac differentiation of P19 mouse embryonic carcinoma stem cells. We found that EOM at 10 µg/ml (a non-cytotoxic dose level) significantly reduced the proportion of cardiac muscle troponin (cTnT) positive cells and the percentage of spontaneously beating embryoid bodies, indicating a severe inhibition of cardiac differentiation. Immunofluorescence and qPCR data demonstrated that EOM increased the expression levels of the aryl hydrocarbon receptor (AhR) and its target gene Cyp1A1 and diminished the expression level of ß-catenin. Furthermore, EOM treatment significantly upregulated cell proliferation rate and elevated the percentage of γH2A.X positive cells without affecting apoptosis. It is worth noting that the EOM-induced changes in gene expression, cellular proliferation and DNA double strain breaks were attenuated by the AhR antagonist CH223191. In conclusion, our data indicate that AhR mediates the inhibitory effects of EOM (from PM2.5) on the cardiac differentiation of P19 cells.


Assuntos
Cardiotoxicidade/tratamento farmacológico , Células-Tronco de Carcinoma Embrionário/metabolismo , MicroRNAs/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Células-Tronco de Carcinoma Embrionário/patologia , Expressão Gênica , Camundongos
18.
Proc Natl Acad Sci U S A ; 115(41): 10434-10439, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30249655

RESUMO

HERV-K HML-2 (HK2) has been proliferating in the germ line of humans at least as recently as 250,000 years ago, with some integrations that remain polymorphic in the modern human population. One of the solitary HK2 LTR polymorphic integrations lies between exons 17 and 18 of RASGRF2, a gene that affects dopaminergic activity and is thus related to addiction. Here we show that this antisense HK2 integration (namely RASGRF2-int) is found more frequently in persons who inject drugs compared with the general population. In a Greek HIV-1-positive population (n = 202), we found RASGRF2-int 2.5 times (14 versus 6%) more frequently in patients infected through i.v. drug use compared with other transmission route controls (P = 0.03). Independently, in a United Kingdom-based hepatitis C virus-positive population (n = 184), we found RASGRF2-int 3.6 times (34 versus 9.5%) more frequently in patients infected during chronic drug abuse compared with controls (P < 0.001). We then tested whether RASGRF2-int could be mechanistically responsible for this association by modulating transcription of RASGRF2 We show that the CRISPR/Cas9-mediated insertion of HK2 in HEK293 cells in the exact RASGRF2 intronic position found in the population resulted in significant transcriptional and phenotypic changes. We also explored mechanistic features of other intronic HK2 integrations and show that HK2 LTRs can be responsible for generation of cis-natural antisense transcripts, which could interfere with the transcription of nearby genes. Our findings suggest that RASGRF2-int is a strong candidate for dopaminergic manipulation, and emphasize the importance of accurate mapping of neglected HERV polymorphisms in human genomic studies.


Assuntos
Células-Tronco de Carcinoma Embrionário/metabolismo , Retrovirus Endógenos/genética , Abuso de Substâncias por Via Intravenosa/genética , Transcrição Gênica , Integração Viral/genética , Fatores ras de Troca de Nucleotídeo Guanina/genética , Estudos de Casos e Controles , Criança , Estudos de Coortes , Células-Tronco de Carcinoma Embrionário/patologia , Feminino , Genoma Humano , Humanos , Masculino , Células Tumorais Cultivadas
19.
Cell Death Dis ; 9(10): 990, 2018 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-30250195

RESUMO

Cancer cells with a less differentiated stem-like phenotype are more resistant to therapeutic manipulations than their differentiated counterparts, and are considered as one of the main causes of cancer persistence and relapse. As such, induction of differentiation in cancer stem-like cells (CSLCs) has emerged as an alternative strategy to enhance the efficacy of anticancer therapies. CSLCs are metabolically distinct from differentiated cells, and any aberration from the intrinsic metabolic state can induce differentiation of CSLCs. Therefore, metabolism-related molecular targets, with a capacity to promote differentiation within CSLCs, are of therapeutic importance. Here, we demonstrate that phosphoglycerate dehydrogenase (PHGDH), an essential enzyme catalyzing the synthesis of amino acid serine, is important for maintaining the poorly differentiated, stem-like state of CSLCs. Our data shows that PHGDH deficiency impairs the tumorsphere formation capacity in embryonal carcinoma stem-like cells (ECSLCs), breast cancer stem-like cells (BCSLCs) and patient-derived brain tumor-initiating cells (BTICs), which is accompanied by the reduced expression of characteristic stemness-promoting factors, such as Oct4, Nanog, Sox-2, and Bmi-1. Mechanistically, PHGDH deficiency in ECSLCs promotes differentiation to various lineages via degradation of Oct4 and by increasing the stability of differentiation marker ß3-tubulin. Furthermore, PHGDH inhibition promotes p-mTOR independent but Beclin-1-dependent autophagy, independent of apoptosis. When studied in combination, the inhibition of both PHGDH and p-mTOR in ECSLCs causes further augmentation of autophagy, and additionally promotes apoptosis, demonstrating the clinical applicability of PHGDH-based manipulations in cancer therapies. Recapitulating these in vitro findings in CSLC models, the intratumoral PHGDH expression in patient-derived tumors is positively correlated with the mRNA levels of stemness factors, especially Oct4, and cancer patients co-expressing high levels of PHGDH and Oct4 display significantly lower survival than those with low PHGDH/Oct4 co-expression. Altogether, this study identifies a clinically-relevant role for PHGDH in the regulation of stemness-differentiation axis within CSLCs.


Assuntos
Autofagia , Neoplasias Encefálicas/metabolismo , Carcinoma Embrionário/metabolismo , Diferenciação Celular , Células-Tronco de Carcinoma Embrionário/metabolismo , Glioblastoma/metabolismo , Fosfoglicerato Desidrogenase/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Neoplasias Testiculares/metabolismo , Proteína Beclina-1/metabolismo , Neoplasias Encefálicas/patologia , Erros Inatos do Metabolismo dos Carboidratos/metabolismo , Carcinoma Embrionário/patologia , Linhagem Celular Tumoral , Glioblastoma/patologia , Humanos , Masculino , Microcefalia/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fosfoglicerato Desidrogenase/antagonistas & inibidores , Fosfoglicerato Desidrogenase/deficiência , Fosfoglicerato Desidrogenase/genética , Proteólise , Transtornos Psicomotores/metabolismo , Convulsões/metabolismo , Neoplasias Testiculares/patologia , Transfecção , Tubulina (Proteína)/metabolismo , Ubiquitinação
20.
In Vitro Cell Dev Biol Anim ; 54(8): 600-609, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30112697

RESUMO

Uc.40 is a long noncoding RNA that is highly conserved among different species, although its function is unknown. It is highly expressed in abnormal human embryonic heart. We previously reported that overexpression of uc.40 promoted apoptosis and inhibited proliferation of P19 cells, and downregulated PBX1, which was identified as a potential target gene of uc.40. The current study evaluated the effects of uc40-siRNA-44 (siRNA against uc.40) on the differentiation, proliferation, apoptosis, and mitochondrial function in P19 cells, and investigated the relationship between uc.40 and PBX1 in cardiomyocytes. The uc.40 silencing expression was confirmed by quantitative real-time polymerase chain reaction (RT-PCR). Observation of morphological changes in transfected P19 cells during different stages of differentiation revealed that uc40-siRNA-44 increased the number of cardiomyocyes. There was no significant difference in the morphology or time of differentiation between the uc40-siRNA-44 group and the control group. uc40-siRNA-44 significantly promoted proliferation of P19 cells and inhibited serum starvation-induced apoptosis. There was no significant difference in mitochondrial DNA copy number or cellular ATP level between the two groups, and ROS levels were significantly decreased in uc40-siRNA-44-transfected cells. The levels of PBX1 and myocardial markers of differentiation were examined in transfected P19 cells; uc40-siRNA-44 downregulated myocardial markers and upregulated PBX1 expression. These results suggest that uc.40 may play an important role during the differentiation of P19 cells by regulation of PBX1 to promote proliferation and inhibit apoptosis. These studies provide a foundation for further study of uc.40/PBX1 in cardiac development.


Assuntos
Diferenciação Celular , Células-Tronco de Carcinoma Embrionário/citologia , Células-Tronco de Carcinoma Embrionário/metabolismo , Inativação Gênica , Miócitos Cardíacos/citologia , Fator de Transcrição 1 de Leucemia de Células Pré-B/genética , RNA Longo não Codificante/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/genética , Biomarcadores/metabolismo , Diferenciação Celular/genética , Proliferação de Células , DNA Mitocondrial/genética , Regulação para Baixo/genética , Metaloproteinases da Matriz/metabolismo , Camundongos , Mitocôndrias/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Especificidade de Órgãos , Fator de Transcrição 1 de Leucemia de Células Pré-B/metabolismo , RNA Longo não Codificante/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transfecção , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...