Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Cell Rep ; 39(7): 110818, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35584683

RESUMO

Histone deacetylases (HDACs) are a class of enzymes that control chromatin state and influence cell fate. We evaluated the chromatin accessibility and transcriptome dynamics of zinc-containing HDACs during cell differentiation in vitro coupled with chemical perturbation to identify the role of HDACs in mesendoderm cell fate specification. Single-cell RNA sequencing analyses of HDAC expression during human pluripotent stem cell (hPSC) differentiation in vitro and mouse gastrulation in vivo reveal a unique association of HDAC1 and -3 with mesendoderm gene programs during exit from pluripotency. Functional perturbation with small molecules reveals that inhibition of HDAC1 and -3, but not HDAC2, induces mesoderm while impeding endoderm and early cardiac progenitor specification. These data identify unique biological functions of the structurally homologous enzymes HDAC1-3 in influencing hPSC differentiation from pluripotency toward mesendodermal and cardiac progenitor populations.


Assuntos
Endoderma , Histona Desacetilases , Células-Tronco Pluripotentes , Animais , Diferenciação Celular/genética , Cromatina/metabolismo , Endoderma/citologia , Endoderma/enzimologia , Endoderma/metabolismo , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/genética , Histona Desacetilase 2/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/enzimologia , Células-Tronco Pluripotentes/metabolismo
2.
Cell Reprogram ; 23(4): 221-238, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34227846

RESUMO

Porcine embryonic stem cells (pESCs) would provide potentials for agricultural- and biotechnological-related applications. However, authentic pESCs have not been established yet because standards for porcine stem cell-specific markers and culture conditions are not clear. Therefore, the present study reports attempts to derive pluripotent epiblast stem cells either from in vitro or in vivo derived porcine embryos. Nine epiblast cell lines (seven lines from Berkshire and two lines from Duroc) could only be isolated from day 9- to 9.5-old in vivo derived early conceptuses. Pluripotency features were analyzed in relation to the presence or absence of alkaline phosphatase (AP) activity. Interestingly, the mRNA expression of several marker genes for pluripotency or epiblast was different between putative epiblast stem cells of the two groups [AP-positive (+) pEpiSC-like cell 2 line and AP-negative (-) pEpiSC-like cell 8 line]. For example, expressions of OCT-3/4, NANOG, SOX2, c-MYC, FGF2, and NODAL in AP-negative (-) porcine epiblast stem cell (pEpiSC)-like cells were higher than those in AP-positive (+) pEpiSC-like cells. Expression of surface markers differed between the two groups to some extent. SSEA-1 was strongly expressed only in AP-negative (-) pEpiSC-like cells, whereas AP-positive (+) pEpiSC-like cells did not express. In addition, we report to have some differences in the in vitro differentiation capacity between AP-positive (+) and AP-negative (-) epiblast cell lines. Primary embryonic germ layer markers (cardiac actin, nestin, and GATA 6) and primordial germ cell markers (Dazl and Vasa) were strongly expressed in embryoid bodies (EBs) aggregated from AP-negative (-) pEpiSC-like cells, whereas EBs aggregated from AP-positive (+) pEpiSCs did not show expression of primary embryonic germ layers and primordial germ cell markers except GATA 6. These results indicate that pEpiSC-like cells display different pluripotency characteristics in relation to AP activity.


Assuntos
Fosfatase Alcalina/metabolismo , Diferenciação Celular , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Camadas Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Animais , Embrião de Mamíferos/enzimologia , Corpos Embrioides/citologia , Corpos Embrioides/enzimologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , Feminino , Camadas Germinativas/enzimologia , Células-Tronco Pluripotentes/enzimologia , Suínos
3.
Nature ; 591(7851): 620-626, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33731924

RESUMO

Limited access to embryos has hampered the study of human embryogenesis and disorders that occur during early pregnancy. Human pluripotent stem cells provide an alternative means to study human development in a dish1-7. Recent advances in partial embryo models derived from human pluripotent stem cells have enabled human development to be examined at early post-implantation stages8-14. However, models of the pre-implantation human blastocyst are lacking. Starting from naive human pluripotent stem cells, here we developed an effective three-dimensional culture strategy with successive lineage differentiation and self-organization to generate blastocyst-like structures in vitro. These structures-which we term 'human blastoids'-resemble human blastocysts in terms of their morphology, size, cell number, and composition and allocation of different cell lineages. Single-cell RNA-sequencing analyses also reveal the transcriptomic similarity of blastoids to blastocysts. Human blastoids are amenable to embryonic and extra-embryonic stem cell derivation and can further develop into peri-implantation embryo-like structures in vitro. Using chemical perturbations, we show that specific isozymes of protein kinase C have a critical function in the formation of the blastoid cavity. Human blastoids provide a readily accessible, scalable, versatile and perturbable alternative to blastocysts for studying early human development, understanding early pregnancy loss and gaining insights into early developmental defects.


Assuntos
Blastocisto/citologia , Blastocisto/metabolismo , Diferenciação Celular , Células-Tronco Pluripotentes/citologia , Blastocisto/enzimologia , Técnicas de Cultura de Células/métodos , Linhagem Celular , Linhagem da Célula , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/enzimologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Isoenzimas/metabolismo , Células-Tronco Pluripotentes/enzimologia , Células-Tronco Pluripotentes/metabolismo , Proteína Quinase C/metabolismo , Análise de Célula Única , Transcriptoma
4.
J Mol Cell Biol ; 13(2): 104-115, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33125075

RESUMO

Mast cells (MCs) play a pivotal role in the hypersensitivity reaction by regulating the innate and adaptive immune responses. Humans have two types of MCs. The first type, termed MCTC, is found in the skin and other connective tissues and expresses both tryptase and chymase, while the second, termed MCT, which only expresses tryptase, is found primarily in the mucosa. MCs induced from human adult-type CD34+ cells are reported to be of the MCT type, but the development of MCs during embryonic/fetal stages is largely unknown. Using an efficient coculture system, we identified that a CD34+c-kit+ cell population, which appeared prior to the emergence of CD34+CD45+ hematopoietic stem and progenitor cells (HSPCs), stimulated robust production of pure Tryptase+Chymase+ MCs (MCTCs). Single-cell analysis revealed dual development directions of CD34+c-kit+ progenitors, with one lineage developing into erythro-myeloid progenitors (EMP) and the other lineage developing into HSPC. Interestingly, MCTCs derived from early CD34+c-kit+ cells exhibited strong histamine release and immune response functions. Particularly, robust release of IL-17 suggested that these early developing tissue-type MCTCs could play a central role in tumor immunity. These findings could help elucidate the mechanisms controlling early development of MCTCs and have significant therapeutic implications.


Assuntos
Diferenciação Celular/genética , Quimases/genética , Mastócitos/citologia , Mastócitos/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Triptases/genética , Biomarcadores , Biomarcadores Tumorais , Células Cultivadas , Quimases/metabolismo , Técnicas de Cocultura , Citocinas/biossíntese , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Liberação de Histamina , Humanos , Modelos Biológicos , Fenótipo , Células-Tronco Pluripotentes/enzimologia , Triptases/metabolismo
5.
Biochem Soc Trans ; 47(3): 875-885, 2019 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-31209155

RESUMO

Ten-eleven translocation (TET) methylcytosine dioxygenases (TET1, TET2, TET3) actively cause demethylation of 5-methylcytosine (5mC) and produce and safeguard hypomethylation at key regulatory regions across the genome. This 5mC erasure is particularly important in pluripotent embryonic stem cells (ESCs) as they need to maintain self-renewal capabilities while retaining the potential to generate different cell types with diverse 5mC patterns. In this review, we discuss the multiple roles of TET proteins in mouse ESCs, and other vertebrate model systems, with a particular focus on TET functions in pluripotency, differentiation, and developmental DNA methylome reprogramming. Furthermore, we elaborate on the recently described non-catalytic roles of TET proteins in diverse biological contexts. Overall, TET proteins are multifunctional regulators that through both their catalytic and non-catalytic roles carry out myriad functions linked to early developmental processes.


Assuntos
Desmetilação do DNA , Dioxigenases/metabolismo , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Humanos , Células-Tronco Pluripotentes/enzimologia , Ligação Proteica
6.
Adv Mater ; 31(17): e1805764, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30767289

RESUMO

Developmental biology has advanced the understanding of the intricate and dynamic processes involved in the formation of an organism from a single cell. However, many gaps remain in the knowledge of embryonic development, especially regarding tissue morphogenesis. A possible approach to mimic such phenomena uses pluripotent stem cells in in vitro morphogenetic models. Herein, these systems are summarized with emphasis on the ability to better manipulate and control cellular interfaces with either liquid or solid materials using microengineered tools, which is critical for attaining deeper insights into pattern formation and stem cell differentiation during organogenesis. The role of conventional and customized cell-culture systems in supporting important advances in the field of morphogenesis is discussed, and the fascinating role that material sciences and microengineering currently play and are expected to play in the future is highlighted. In conclusion, it is proffered that continued microfluidics innovations when applied to morphogenesis promise to provide important insights to advance many multidisciplinary fields, including regenerative medicine.


Assuntos
Microfluídica/métodos , Morfogênese/efeitos dos fármacos , Células-Tronco Pluripotentes/enzimologia , Células-Tronco Pluripotentes/metabolismo , Biomimética/métodos , Técnicas de Cultura de Células , Diferenciação Celular , Membrana Celular/enzimologia , Membrana Celular/metabolismo , Humanos , Organogênese , Polímeros/química , Propriedades de Superfície , Alicerces Teciduais/química
7.
Stem Cell Reports ; 11(6): 1347-1356, 2018 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-30503259

RESUMO

Nicotinamide, the amide form of vitamin B3, is widely used in disease treatments and stem cell applications. However, nicotinamide's impact often cannot be attributed to its nutritional functions. In a vitamin screen, we find that nicotinamide promotes cell survival and differentiation in human pluripotent stem cells. Nicotinamide inhibits the phosphorylation of myosin light chain, suppresses actomyosin contraction, and leads to improved cell survival after individualization. Further analysis demonstrates that nicotinamide is an inhibitor of multiple kinases, including ROCK and casein kinase 1. We demonstrate that nicotinamide affects human embryonic stem cell pluripotency and differentiation as a selective kinase inhibitor. The findings in this report may help researchers design better strategies to develop nicotinamide-related stem cell applications and disease treatments.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Niacinamida/farmacologia , Células-Tronco Pluripotentes/citologia , Inibidores de Proteínas Quinases/farmacologia , Actomiosina/metabolismo , Caseína Quinase I/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , NAD/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/enzimologia , Células-Tronco Pluripotentes/metabolismo , Quinases Associadas a rho/metabolismo
8.
Nat Commun ; 9(1): 4815, 2018 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-30446643

RESUMO

Common disorders, including diabetes and Parkinson's disease, are caused by a combination of environmental factors and genetic susceptibility. However, defining the mechanisms underlying gene-environment interactions has been challenging due to the lack of a suitable experimental platform. Using pancreatic ß-like cells derived from human pluripotent stem cells (hPSCs), we discovered that a commonly used pesticide, propargite, induces pancreatic ß-cell death, a pathological hallmark of diabetes. Screening a panel of diverse hPSC-derived cell types we extended this observation to a similar susceptibility in midbrain dopamine neurons, a cell type affected in Parkinson's disease. We assessed gene-environment interactions using isogenic hPSC lines for genetic variants associated with diabetes and Parkinson's disease. We found GSTT1-/- pancreatic ß-like cells and dopamine neurons were both hypersensitive to propargite-induced cell death. Our study identifies an environmental chemical that contributes to human ß-cell and dopamine neuron loss and validates a novel hPSC-based platform for determining gene-environment interactions.


Assuntos
Cicloexanos/toxicidade , Diabetes Mellitus/induzido quimicamente , Neurônios Dopaminérgicos/efeitos dos fármacos , Interação Gene-Ambiente , Células Secretoras de Insulina/efeitos dos fármacos , Praguicidas/toxicidade , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Diferenciação Celular , Diabetes Mellitus/genética , Diabetes Mellitus/patologia , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/enzimologia , Glutationa Transferase/deficiência , Glutationa Transferase/genética , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/enzimologia , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/enzimologia , Camundongos , Modelos Biológicos , Doença de Parkinson/etiologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/enzimologia
9.
Cell Rep ; 24(4): 895-908.e6, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-30044986

RESUMO

The role of primary cilia in mechanosensation is essential in endothelial cell (EC) shear responsiveness. Here, we find that venous, capillary, and progenitor ECs respond to shear stress in vitro in a cilia-dependent manner. We then demonstrate that primary cilia assembly in human induced pluripotent stem cell (hiPSC)-derived ECs varies between different cell lines with marginal influence of differentiation protocol. hiPSC-derived ECs lacking cilia do not align to shear stress, lack stress fiber assembly, have uncoordinated migration during wound closure in vitro, and have aberrant calcium influx upon shear exposure. Transcriptional analysis reveals variation in regulatory genes involved in ciliogenesis among different hiPSC-derived ECs. Moreover, inhibition of histone deacetylase 6 (HDAC6) activity in hiPSC-ECs lacking cilia rescues cilia formation and restores mechanical sensing. Taken together, these results show the importance of primary cilia in hiPSC-EC mechano-responsiveness and its modulation through HDAC6 activity varies among hiPSC-ECs.


Assuntos
Cílios/enzimologia , Células Endoteliais/enzimologia , Desacetilase 6 de Histona/metabolismo , Células-Tronco Pluripotentes/enzimologia , Cálcio/metabolismo , Movimento Celular/fisiologia , Citoesqueleto/enzimologia , Células Endoteliais/citologia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Mecanotransdução Celular , Técnicas Analíticas Microfluídicas , Células-Tronco Pluripotentes/citologia , Artérias Umbilicais/citologia , Artérias Umbilicais/enzimologia
10.
Mech Dev ; 154: 60-63, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29753812

RESUMO

Pluripotent stem cells (PSCs) are capable of self-renewing and producing all cell types derived from the three germ layers in response to developmental cues, constituting an important promise for regenerative medicine. Pluripotency depends on specific transcription factors (TFs) that induce genes required to preserve the undifferentiated state and repress other genes related to differentiation. The transcription machinery and regulatory components such as TFs are recruited dynamically on their target genes making it essential exploring their dynamics in living cells to understand the transcriptional output. Non-invasive and very sensitive fluorescence microscopy methods are making it possible visualizing the dynamics of TFs in living specimens, complementing the information extracted from studies in fixed specimens and bulk assays. In this work, we briefly describe the basis of these microscopy methods and review how they contributed to our knowledge of the function of TFs relevant to embryo development and cell differentiation in a variety of systems ranging from single cells to whole organisms.


Assuntos
Desenvolvimento Embrionário/fisiologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/fisiologia , Camadas Germinativas/embriologia , Camadas Germinativas/metabolismo , Humanos , Microscopia de Fluorescência/métodos , Células-Tronco Pluripotentes/enzimologia , Células-Tronco Pluripotentes/metabolismo , Transcrição Gênica/fisiologia
11.
Cell Stem Cell ; 21(4): 449-455.e4, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28985526

RESUMO

Mouse embryonic stem cells (ESCs) cultured in serum are characterized by hyper-phosphorylated RB protein, lack of G1 control, and rapid progression through the cell cycle. Here, we show that ESCs grown in the presence of two small-molecule inhibitors (2i ESCs) have a longer G1-phase with hypo-phosphorylated RB, implying that they have a functional G1 checkpoint. Deletion of RB, P107, and P130 in 2i ESCs results in a G1-phase similar to that of serum ESCs. Inhibition of the ERK signaling pathway in serum ESCs results in the appearance of hypo-phosphorylated RB and the reinstatement of a G1 checkpoint. In addition, induction of a dormant state by the inhibition of MYC, resembling diapause, requires the presence of the RB family proteins. Collectively, our data show that RB-dependent G1 restriction point signaling is active in mouse ESCs grown in 2i but abrogated in serum by ERK-dependent phosphorylation.


Assuntos
Ciclo Celular , Células-Tronco Pluripotentes/citologia , Animais , Fase G1 , Sistema de Sinalização das MAP Quinases , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/enzimologia , Células-Tronco Pluripotentes/enzimologia , Proteína do Retinoblastoma/metabolismo
12.
Nat Cell Biol ; 19(7): 763-773, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28604677

RESUMO

Naive pluripotency is established in preimplantation epiblast. Embryonic stem cells (ESCs) represent the immortalization of naive pluripotency. 2i culture has optimized this state, leading to a gene signature and DNA hypomethylation closely comparable to preimplantation epiblast, the developmental ground state. Here we show that Pramel7 (PRAME-like 7), a protein highly expressed in the inner cell mass (ICM) but expressed at low levels in ESCs, targets for proteasomal degradation UHRF1, a key factor for DNA methylation maintenance. Increasing Pramel7 expression in serum-cultured ESCs promotes a preimplantation epiblast-like gene signature, reduces UHRF1 levels and causes global DNA hypomethylation. Pramel7 is required for blastocyst formation and its forced expression locks ESCs in pluripotency. Pramel7/UHRF1 expression is mutually exclusive in ICMs whereas Pramel7-knockout embryos express high levels of UHRF1. Our data reveal an as-yet-unappreciated dynamic nature of DNA methylation through proteasome pathways and offer insights that might help to improve ESC culture to reproduce in vitro the in vivo ground-state pluripotency.


Assuntos
Antígenos de Neoplasias/metabolismo , Blastocisto/enzimologia , Células-Tronco Embrionárias/enzimologia , Epigênese Genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Células-Tronco Pluripotentes/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Antígenos de Neoplasias/genética , Blastocisto/citologia , Proteínas Estimuladoras de Ligação a CCAAT , Proteínas Culina/metabolismo , Metilação de DNA , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Fenótipo , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Proteólise , Interferência de RNA , Fatores de Tempo , Transcriptoma , Transfecção , Ubiquitina-Proteína Ligases
13.
Elife ; 62017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28654420

RESUMO

During early embryogenesis, cells must exit pluripotency and commit to multiple lineages in all germ-layers. How this transition is operated in vivo is poorly understood. Here, we report that MEK1 and the Nanog-related transcription factor Ventx2 coordinate this transition. MEK1 was required to make Xenopus pluripotent cells competent to respond to all cell fate inducers tested. Importantly, MEK1 activity was necessary to clear the pluripotency protein Ventx2 at the onset of gastrulation. Thus, concomitant MEK1 and Ventx2 knockdown restored the competence of embryonic cells to differentiate. Strikingly, MEK1 appeared to control the asymmetric inheritance of Ventx2 protein following cell division. Consistently, when Ventx2 lacked a functional PEST-destruction motif, it was stabilized, displayed symmetric distribution during cell division and could efficiently maintain pluripotency gene expression over time. We suggest that asymmetric clearance of pluripotency regulators may represent an important mechanism to ensure the progressive assembly of primitive embryonic tissues.


Assuntos
Diferenciação Celular , Proteínas de Homeodomínio/metabolismo , MAP Quinase Quinase 1/metabolismo , Células-Tronco Pluripotentes/enzimologia , Células-Tronco Pluripotentes/fisiologia , Fatores de Transcrição/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus/embriologia , Animais
14.
Stem Cells ; 35(9): 2050-2059, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28631381

RESUMO

Human pluripotent stem cells (hPSC) require signaling provided by fibroblast growth factor (FGF) receptors. This can be initiated by the recombinant FGF2 ligand supplied exogenously, but hPSC further support their niche by secretion of endogenous FGF2. In this study, we describe a role of tyrosine kinase expressed in hepatocellular carcinoma (TEC) kinase in this process. We show that TEC-mediated FGF2 secretion is essential for hPSC self-renewal, and its lack mediates specific differentiation. Following both short hairpin RNA- and small interfering RNA-mediated TEC knockdown, hPSC secretes less FGF2. This impairs hPSC proliferation that can be rescued by increasing amounts of recombinant FGF2. TEC downregulation further leads to a lower expression of the pluripotency markers, an improved priming towards neuroectodermal lineage, and a failure to develop cardiac mesoderm. Our data thus demonstrate that TEC is yet another regulator of FGF2-mediated hPSC pluripotency and differentiation. Stem Cells 2017;35:2050-2059.


Assuntos
Linhagem da Célula , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/enzimologia , Proteínas Tirosina Quinases/metabolismo , Biomarcadores/metabolismo , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Proteínas Recombinantes/farmacologia
15.
J Mol Biol ; 429(10): 1504-1520, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28456524

RESUMO

Post-translational modification of proteins by phosphorylation plays a key role in regulating all aspects of eukaryotic biology. Embryonic stem cell (ESC) pluripotency, defined as the ability to differentiate into all cell types in the adult body, is no exception. Maintenance and dissolution of pluripotency are tightly controlled by phosphorylation. As a result, key signalling pathways that regulate pluripotency have been identified and their functions well characterised. Amongst the best studied are the fibroblast growth factor (FGF)-ERK1/2 pathway, PI3K-AKT, the leukemia inhibitory factor (LIF)-JAK-STAT3 axis, Wnt-GSK3 signalling, and the transforming growth factor (TGF)ß family. However, these kinase pathways constitute only a small proportion of the protein kinase complement of pluripotent cells, and there is accumulating evidence that diverse phosphorylation systems modulate ESC pluripotency. Here, we review recent progress in understanding the overarching role of phosphorylation in mediating communication from the cellular environment, metabolism, and cell cycle to the core pluripotency machinery.


Assuntos
Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/fisiologia , Células-Tronco Pluripotentes/enzimologia , Células-Tronco Pluripotentes/fisiologia , Proteínas Quinases/metabolismo , Transdução de Sinais , Animais , Regulação da Expressão Gênica , Humanos , Camundongos , Fosforilação , Processamento de Proteína Pós-Traducional
16.
Nat Cell Biol ; 19(5): 445-456, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28436968

RESUMO

A hallmark of cancer cells is the metabolic switch from oxidative phosphorylation (OXPHOS) to glycolysis, a phenomenon referred to as the 'Warburg effect', which is also observed in primed human pluripotent stem cells (hPSCs). Here, we report that downregulation of SIRT2 and upregulation of SIRT1 is a molecular signature of primed hPSCs and that SIRT2 critically regulates metabolic reprogramming during induced pluripotency by targeting glycolytic enzymes including aldolase, glyceraldehyde-3-phosphate dehydrogenase, phosphoglycerate kinase, and enolase. Remarkably, knockdown of SIRT2 in human fibroblasts resulted in significantly decreased OXPHOS and increased glycolysis. In addition, we found that miR-200c-5p specifically targets SIRT2, downregulating its expression. Furthermore, SIRT2 overexpression in hPSCs significantly affected energy metabolism, altering stem cell functions such as pluripotent differentiation properties. Taken together, our results identify the miR-200c-SIRT2 axis as a key regulator of metabolic reprogramming (Warburg-like effect), via regulation of glycolytic enzymes, during human induced pluripotency and pluripotent stem cell function.


Assuntos
Diferenciação Celular , Metabolismo Energético , MicroRNAs/metabolismo , Células-Tronco Pluripotentes/enzimologia , Sirtuína 2/metabolismo , Acetilação , Linhagem da Célula , Sobrevivência Celular , Reprogramação Celular , Biologia Computacional , Bases de Dados Genéticas , Regulação Enzimológica da Expressão Gênica , Glicólise , Células HEK293 , Humanos , MicroRNAs/genética , Fosforilação Oxidativa , Fenótipo , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/metabolismo , Sirtuína 2/genética , Fatores de Tempo , Transfecção
17.
Mol Cell ; 65(4): 589-603.e9, 2017 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-28212747

RESUMO

Pioneer transcription factors (TFs) function as genomic first responders, binding to inaccessible regions of chromatin to promote enhancer formation. The mechanism by which pioneer TFs gain access to chromatin remains an important unanswered question. Here we show that PARP-1, a nucleosome-binding protein, cooperates with intrinsic properties of the pioneer TF Sox2 to facilitate its binding to intractable genomic loci in embryonic stem cells. These actions of PARP-1 occur independently of its poly(ADP-ribosyl) transferase activity. PARP-1-dependent Sox2-binding sites reside in euchromatic regions of the genome with relatively high nucleosome occupancy and low co-occupancy by other transcription factors. PARP-1 stabilizes Sox2 binding to nucleosomes at suboptimal sites through cooperative interactions on DNA. Our results define intrinsic and extrinsic features that determine Sox2 pioneer activity. The conditional pioneer activity observed with Sox2 at a subset of binding sites may be a key feature of other pioneer TFs operating at intractable genomic loci.


Assuntos
DNA/metabolismo , Células-Tronco Embrionárias/enzimologia , Eucromatina/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Loci Gênicos , Nucleossomos/enzimologia , Células-Tronco Pluripotentes/enzimologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , DNA/genética , Eucromatina/genética , Humanos , Camundongos , Nucleossomos/genética , Poli(ADP-Ribose) Polimerase-1/genética , Ligação Proteica , Fatores de Transcrição SOXB1/genética , Transdução de Sinais , Fatores de Tempo , Transfecção
18.
PLoS One ; 11(10): e0163244, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27723793

RESUMO

Multiple pluripotent cell populations, which together comprise the pluripotent cell lineage, have been identified. The mechanisms that control the progression between these populations are still poorly understood. The formation of early primitive ectoderm-like (EPL) cells from mouse embryonic stem (mES) cells provides a model to understand how one such transition is regulated. EPL cells form from mES cells in response to l-proline uptake through the transporter Slc38a2. Using inhibitors of cell signaling we have shown that Src family kinases, p38 MAPK, ERK1/2 and GSK3ß are required for the transition between mES and EPL cells. ERK1/2, c-Src and GSK3ß are likely to be enforcing a receptive, primed state in mES cells, while Src family kinases and p38 MAPK are involved in the establishment of EPL cells. Inhibition of these pathways prevented the acquisition of most, but not all, features of EPL cells, suggesting that other pathways are required. L-proline activation of differentiation is mediated through metabolism and changes to intracellular metabolite levels, specifically reactive oxygen species. The implication of multiple signaling pathways in the process suggests a model in which the context of Src family kinase activation determines the outcomes of pluripotent cell differentiation.


Assuntos
Diferenciação Celular , Sistema de Sinalização das MAP Quinases , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Pluripotentes/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Quinases da Família src/metabolismo , Animais , Técnicas de Cultura de Células , Ativação Enzimática , Camundongos , Espécies Reativas de Oxigênio/metabolismo
19.
Stem Cells Dev ; 25(14): 1050-9, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27216987

RESUMO

The discovery of mammalian N(6)-methyladenosine (m(6)A) methyltransferases and demethylases has enriched our knowledge of the dynamic regulation of the most prevalent posttranscriptional RNA modification, m(6)A methylation. This reversible methylation process of adding and removing m(6)A marks on RNA has been shown to have broad biological functions in fine tuning cellular processes and gene expression. Recent studies have revealed a critical role for the currently known m(6)A methyltransferases and demethylases in regulating the pluripotency and differentiation of stem cells. These data establish a novel dimension in epigenetic regulation at the RNA level to affect mammalian cell fate.


Assuntos
Adenosina/análogos & derivados , Diferenciação Celular , Metiltransferases/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/enzimologia , Adenosina/metabolismo , Animais , Humanos , RNA/metabolismo
20.
Sci Rep ; 6: 22190, 2016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-26911679

RESUMO

Epigenomic regulation is likely to be important in the maintenance of genomic integrity of human pluripotent stem cells, however, the mechanisms are unknown. We explored the epigenomes and transcriptomes of human pluripotent stem cells before and after spontaneous transformation to abnormal karyotypes and in correlation to cancer cells. Our results reveal epigenetic silencing of Catalase, a key regulator of oxidative stress and DNA damage control in abnormal cells. Our findings provide novel insight into the mechanisms associated with spontaneous transformation of human pluripotent stem cells towards malignant fate. The same mechanisms may control the genomic stability of cells in somatic tissues.


Assuntos
Cariótipo Anormal , Catalase/genética , Inativação Gênica , Células-Tronco Pluripotentes/metabolismo , Neoplasias Testiculares/genética , Estudos de Casos e Controles , Catalase/metabolismo , Linhagem Celular , Humanos , Masculino , Estresse Oxidativo , Células-Tronco Pluripotentes/enzimologia , Neoplasias Testiculares/metabolismo , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA