Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Stem Cells ; 40(3): 239-259, 2022 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-35323987

RESUMO

Metabolism plays a crucial role for cell survival and function; however, recent evidence has implicated it in regulating embryonic development. In the embryo, the inner cell mass undergoes orchestrated cellular divisions resulting in the formation of pluripotent epiblast stem cells and primitive endoderm cells. However, both lineages can be captured in vitro as embryonic stem (ES) cells and extraembryonic endoderm (XEN) cells. Concomitantly, changes in the metabolic profile occurs during development, and are well documented in the embryonic lineages. However, a comprehensive multi-omic analysis of these features in XEN cells remains lacking. We observed that mouse XEN cells exhibited high sensitivity to glycolytic inhibition in addition to maintaining elevated intra- and extracellular lactate levels in vitro. Extraembryonic endoderm cells maintain high lactate levels by increased LDHA activity, and re-routing pyruvate away from the mitochondria resulting in reduced mitochondrial activity due to disruptions in electron transport chain stoichiometry. Importantly, exogenous lactate supplementation or promoting intracellular lactate accumulation enhances XEN differentiation in vitro. These results highlight how lactate contributes to XEN differentiation in vitro and may serve to enhance reprogramming efficiency of cells used for regenerative medicine.


Assuntos
Endoderma , Ácido Láctico , Animais , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Ácido Láctico/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas
2.
Biochem Cell Biol ; 97(5): 600-611, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30880404

RESUMO

PARP2 belongs to a family of proteins involved in cell differentiation, DNA damage repair, cellular energy expenditure, and chromatin modeling. In addition to these overlapping functions with PARP1, PARP2 participates in spermatogenesis, T-cell maturation, extra-embryonic endoderm formation, adipogenesis, lipid metabolism, and cholesterol homeostasis. Knowledge of the functions of PARP2 is far from complete, and the mechanism(s) by which the gene and protein are regulated are unknown. In this study, we found that two different mechanisms are used in vitro to regulate PARP2 levels. In the presence of serum, PARP2 is degraded through the ubiquitin-proteasome pathway; however, when serum is removed or dialyzed with a 3.5 kDa molecular cut membrane, PARP2 rapidly becomes sodium dodecyl sulphate- and urea-insoluble. Despite the presence of a putative serum response element in the PARP2 gene, transcription is not affected by serum deprivation, and PARP2 levels are restored when serum is replaced. The loss of PARP2 affects cell differentiation and gene expression linked to cholesterol and lipid metabolism. These observations highlight the critical roles that PARP2 plays under different physiological conditions, and reveal that PARP2 is tightly regulated by distinct pathways.


Assuntos
Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos , Poli(ADP-Ribose) Polimerases/sangue , Poli(ADP-Ribose) Polimerases/genética
3.
Biochem Cell Biol ; 95(2): 251-262, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28177772

RESUMO

Mouse F9 cells differentiate into primitive endoderm (PrE) following the activation of the canonical WNT-ß-catenin pathway. The upregulation of Wnt6 and activation of ß-catenin-TCF-LEF-dependent transcription is known to accompany differentiation, but the Frizzled (FZD) receptor responsible for transducing the WNT6 signal is not known. Eight of the 10 Fzd genes were found to be expressed in F9 cells, with Fzd7 being the most highly expressed, and chosen for further analysis. To alter steady-state Fzd7 levels and test the effect this has on differentiation, siRNA and overexpression approaches were used to knock-down and ectopically express the Fzd7 message, respectively. siRNA knock-down of Fzd7 resulted in reduced DAB2 levels, and the overexpression activated a TCF-LEF reporter, but neither approach affected differentiation. Our focus turned to how canonical WNT6 signaling was attenuated to allow PrE cells to form parietal endoderm (PE). Dkk1, encoding a WNT antagonist, was examined and results showed that its expression increased in F9 cells treated with retinoic acid (RA) or overexpressing Wnt6. F9 cells overexpressing human DKK1 or treated with DKK1-conditioned medium and then treated with RA failed to differentiate, indicating that a negative feedback loop involving WNT6 and DKK1 attenuates canonical WNT-ß-catenin signaling, thereby allowing PE cells to differentiate.


Assuntos
Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas Proto-Oncogênicas/genética , Receptores Acoplados a Proteínas G/genética , Teratocarcinoma/genética , Proteínas Wnt/genética , beta Catenina/genética , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Endoderma/metabolismo , Endoderma/patologia , Retroalimentação Fisiológica , Receptores Frizzled , Genes Reporter , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Teratocarcinoma/metabolismo , Teratocarcinoma/patologia , Tretinoína/farmacologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
4.
Stem Cells Dev ; 31(23-24): 741-755, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36103394

RESUMO

Hedgehog signaling is essential for vertebrate development; however, less is known about the negative regulators that influence this pathway. Using the mouse P19 embryonal carcinoma cell model, suppressor of fused (SUFU), a negative regulator of the Hedgehog (Hh) pathway, was investigated during retinoic acid (RA)-induced neural differentiation. We found Hh signaling increased activity in the early phase of differentiation, but was reduced during terminal differentiation of neurons and astrocytes. This early increase in pathway activity was required for neural differentiation; however, it alone was not sufficient to induce neural lineages. SUFU, which regulates signaling at the level of Gli, remained relatively unchanged during differentiation, but its loss through CRISPR-Cas9 gene editing resulted in ectopic expression of Hh target genes. Interestingly, these SUFU-deficient cells were unable to differentiate toward neural lineages without RA, and when directed toward these lineages, they showed delayed and decreased astrocyte differentiation; neuron differentiation was unaffected. Ectopic activation of Hh target genes in SUFU-deficient cells remained throughout RA-induced differentiation and this was accompanied by the loss of Gli3, despite the presence of the Gli3 message. Thus, the study indicates the proper timing and proportion of astrocyte differentiation requires SUFU, likely acting through Gli3, to reduce Hh signaling during late-stage differentiation.


Assuntos
Astrócitos , Proteínas Hedgehog , Proteínas Repressoras , Animais , Camundongos , Diferenciação Celular/genética , Proteínas Hedgehog/genética , Astrócitos/citologia , Transdução de Sinais , Proteínas Repressoras/genética
5.
Cell Signal ; 100: 110484, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36195199

RESUMO

Wnt and Hh are known signalling pathways involved in neural differentiation and recent work has shown the cell cycle regulator, Never in Mitosis Kinase 2 (Nek2) is able to regulate both pathways. Despite its known function in pathway regulation, few studies have explored Nek2 within embryonic development. The P19 embryonal carcinoma cell model was used to investigate Nek2 and neural differentiation through CRISPR knockout and overexpression studies. Loss of Nek2 reduced cell proliferation in the undifferentiated state and during directed differentiation, while overexpression increased cell proliferation. Despite these changes in proliferation rates, Nek2 deficient cells maintained pluripotency markers after neural induction while Nek2 overexpressing cells lost these markers in the undifferentiated state. Nek2 deficient cells lost the ability to differentiate into both neurons and astrocytes, although Nek2 overexpressing cells enhanced neuron differentiation at the expense of astrocytes. Hh and Wnt signalling were explored, however there was no clear connection between Nek2 and these pathways causing the observed changes to differentiation phenotypes. Mass spectrometry was also used during wildtype and Nek2 knockout cell differentiation and we identified reduced electron transport chain components in the knockout population. Immunoblotting confirmed the loss of these components and additional studies showed cells lacking Nek2 were exclusively glycolytic. Interestingly, hypoxia inducible factor 1α was stabilized in these Nek2 knockout cells despite culturing them under normoxic conditions. Since neural differentiation requires a metabolic switch from glycolysis to oxidative phosphorylation, we propose a mechanism where Nek2 prevents HIF1α stabilization, thereby allowing cells to use oxidative phosphorylation to facilitate neuron and astrocyte differentiation.

6.
Biomolecules ; 12(5)2022 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-35625551

RESUMO

The regulation of proteins through the addition and removal of O-linked ß-N-acetylglucosamine (O-GlcNAc) plays a role in many signaling events, specifically in stem cell pluripotency and the regulation of differentiation. However, these post-translational modifications have not been explored in extraembryonic endoderm (XEN) differentiation. Of the plethora of proteins regulated through O-GlcNAc, we explored galectin-3 as a candidate protein known to have various intracellular and extracellular functions. Based on other studies, we predicted a reduction in global O-GlcNAcylation levels and a distinct galectin expression profile in XEN cells relative to embryonic stem (ES) cells. By conducting dot blot analysis, XEN cells had decreased levels of global O-GlcNAc than ES cells, which reflected a disbalance in the expression of genes encoding O-GlcNAc cycle enzymes. Immunoassays (Western blot and ELISA) revealed that although XEN cells (low O-GlcNAc) had lower concentrations of both intracellular and extracellular galectin-3 than ES cells (high O-GlcNAc), the relative secretion of galectin-3 was significantly increased by XEN cells. Inducing ES cells toward XEN in the presence of an O-GlcNAcase inhibitor was not sufficient to inhibit XEN differentiation. However, global O-GlcNAcylation was found to decrease in differentiated cells and the extracellular localization of galectin-3 accompanies these changes. Inhibiting global O-GlcNAcylation status does not, however, impact pluripotency and the ability of ES cells to differentiate to the XEN lineage.


Assuntos
Endoderma , Galectina 3 , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias , Endoderma/metabolismo , Galectina 3/metabolismo , Galectinas/metabolismo
7.
Cancer Res ; 82(7): 1208-1221, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35149587

RESUMO

G9a and EZH2 are two histone methyltransferases commonly upregulated in several cancer types, yet the precise roles that these enzymes play cooperatively in cancer is unclear. We demonstrate here that frequent concurrent upregulation of both G9a and EZH2 occurs in several human tumors. These methyltransferases cooperatively repressed molecular pathways responsible for tumor cell death. In genetically distinct tumor subtypes, concomitant inhibition of G9a and EZH2 potently induced tumor cell death, highlighting the existence of tumor cell survival dependency at the epigenetic level. G9a and EZH2 synergistically repressed expression of genes involved in the induction of endoplasmic reticulum (ER) stress and the production of reactive oxygen species. IL24 was essential for the induction of tumor cell death and was identified as a common target of G9a and EZH2. Loss of function of G9a and EZH2 activated the IL24-ER stress axis and increased apoptosis in cancer cells while not affecting normal cells. These results indicate that G9a and EZH2 promotes the evasion of ER stress-mediated apoptosis by repressing IL24 transcription, therefore suggesting that their inhibition may represent a potential therapeutic strategy for solid cancers. SIGNIFICANCE: These findings demonstrate a novel role for G9a and EZH2 histone methyltransferases in suppressing apoptosis, which can be targeted with small molecule inhibitors as a potential approach to improve solid cancer treatment.


Assuntos
Histona-Lisina N-Metiltransferase , Neoplasias , Apoptose/genética , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Antígenos de Histocompatibilidade/genética , Antígenos de Histocompatibilidade/metabolismo , Histona Metiltransferases/genética , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética
8.
Arch Biochem Biophys ; 516(2): 173-81, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22056482

RESUMO

The gp91phox subunit of flavocytochrome b(558) is the catalytic core of the phagocyte plasma membrane NADPH oxidase. Its activation occurs within lipid rafts and requires translocation of four subunits to flavocytochrome b(558). gp91phox is the only glycosylated subunit of NADPH oxidase and no data exist about the structure or function of its glycans. Glycans, however, bind to lectins and this can stimulate NADPH oxidase activity. Given this information, we hypothesized that lectin-gp91phox interactions would facilitate the assembly of a functionally active NADPH oxidase in the absence of lipid rafts. To test this, we used lectins with different carbohydrate-binding specificity to examine the effects on H(2)O(2) generation by human neutrophils treated with the lipid raft disrupting agent methyl-ß-cyclodextrin (MßCD). MßCD treatment removed membrane cholesterol, caused changes in cell morphology, inhibited lectin-induced cell aggregation, and delayed lectin-induced assembly of the NADPH oxidase complex. More importantly, MßCD treatment either stimulated or inhibited H(2)O(2) production in a lectin-dependent manner. Together, these results show selectivity in lectin binding to gp91phox, and provide evidence for the biochemical structures of the gp91phox glycans. Furthermore, the data also indicate that in the absence of lipid rafts, neutrophil NADPH oxidase activity can be altered by these select lectins.


Assuntos
Colesterol/metabolismo , Lectinas/farmacologia , NADPH Oxidases/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Agregação Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Forma Celular/efeitos dos fármacos , Grupo dos Citocromos b/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/metabolismo , Técnicas In Vitro , Glicoproteínas de Membrana/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Microscopia Eletrônica de Varredura , Modelos Biológicos , NADPH Oxidase 2 , beta-Ciclodextrinas/farmacologia
9.
Clin Cancer Res ; 27(9): 2624-2635, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33589432

RESUMO

PURPOSE: G9a histone methyltransferase exerts oncogenic effects in several tumor types and its inhibition promotes anticancer effects. However, the impact on checkpoint inhibitor blockade response and the utility of G9a or its target genes as a biomarker is poorly studied. We aimed to examine whether G9a inhibition can augment the efficacy of checkpoint inhibitor blockade and whether LC3B, a G9a target gene, can predict treatment response. EXPERIMENTAL DESIGN: Clinical potential of LC3B as a biomarker of checkpoint inhibitor blockade was assessed using patient samples including tumor biopsies and circulating tumor cells from liquid biopsies. Efficacy of G9a inhibition to enhance checkpoint inhibitor blockade was examined using a mouse model. RESULTS: Patients with melanoma who responded to checkpoint inhibitor blockade were associated with not only a higher level of tumor LC3B but also a higher proportion of cells expressing LC3B. A higher expression of MAP1LC3B or LC3B protein was associated with longer survival and lower incidence of acquired resistance to checkpoint inhibitor blockade, suggesting LC3B as a potential predictive biomarker. We demonstrate that G9a histone methyltransferase inhibition is able to not only robustly induce LC3B level to augment the efficacy of checkpoint inhibitor blockade, but also induces melanoma cell death. CONCLUSIONS: Checkpoint inhibitor blockade response is limited to a subset of the patient population. These results have implications for the development of LC3B as a predictive biomarker of checkpoint inhibitor blockade to guide patient selection, as well as G9a inhibition as a strategy to extend the proportion of patients responding to immunotherapy.


Assuntos
Inibidores Enzimáticos/farmacologia , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Inibidores de Checkpoint Imunológico/farmacologia , Melanoma/metabolismo , Animais , Biomarcadores Tumorais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Sinergismo Farmacológico , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Antígenos de Histocompatibilidade/genética , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Melanoma/diagnóstico , Melanoma/tratamento farmacológico , Melanoma/genética , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Células Neoplásicas Circulantes , Prognóstico , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Biol Cell ; 101(11): 661-78, 4 p following 678, 2009 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-19515017

RESUMO

BACKGROUND INFORMATION: PTEN (phosphatase and tensin homologue deleted on chromosome 10) is a negative regulator of the PI3K (phosphoinositide 3-kinase)-Akt (also called protein kinase B) signalling pathway and is essential for embryogenesis, but its function in early vertebrate embryos is unclear. RESULTS: To address how PTEN functions in early embryos, we overexpressed one of the four zebrafish PTEN isoforms at the 1-2-cell stage. Overexpression of Ptena454 alters phospho-Akt levels and impairs cell movements associated with gastrulation. Heat shocking embryos increases phospho-Akt levels and lowers phospho-Ptena454 levels. Inhibiting CK2 (protein kinase CK2) activity reduces phospho-Pten levels and augments the effects due to Ptena454 overexpression. Low phospho-Akt and corresponding low phospho-GSK-3 (glycogen synthase kinase-3) and high phospho-Pten levels accompany wortmannin or LY294002 treatment, which inhibit PI3K activity. CONCLUSIONS: These results suggest that Ptena454 regulation is correlated to changes in phospho-Akt levels. We propose a model in which homoeostasis in rapidly dividing and migrating embryonic cells depends on a counterbalance between pro-survival signalling employing CK2 and GSK-3 and the pro-apoptotic activity of Ptena454.


Assuntos
Gastrulação , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Feminino , Masculino , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
11.
Bone ; 136: 115365, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32320893

RESUMO

Oculodentodigital dysplasia (ODDD) is a disease caused by mutations in the GJA1 gene that encodes the gap-junctional protein connexin43 (Cx43). ODDD affects multiple organs, but craniofacial anomalies are typical. However, details on the timing of phenotypic presentation of these abnormalities and their correspondence with potential cellular changes are incomplete. Here, we perform the first assessment of the development of the ODDD craniofacial phenotype in the Cx43I130T/+ mouse model and show that the phenotypic features commonly found in patients with the disorder arise in mice between E17.5 and birth and become more profound with age. Using mice heterozygous for the I130T mutation of Gja1, we provide a detailed analysis of the craniofacial phenotype in this ODDD model using shape analyses based on micro-CT images. Results show that in addition to differences in facial bone morphology, there are significant shape differences in the cranial base. Mutant mice display delayed ossification at E17.5 and birth, particularly in bones of the face and cranial vault but ossification is normal at three months. Our immunohistochemical analyses of the palatine bone indicate that osteoblast differentiation is delayed in Cx43I130T/+ mice compared to their wildtype littermates, which likely contributes to the phenotypic variations observed in the facial bones. Our histological and immunohistochemical analyses of the synchondroses of the cranial base show no differences in molecular indicators of chondrocyte differentiation in mutant mice, suggesting that the differences to cranial base morphology displayed by Cx43I130T/+ mice are not due to differences in chondrocyte proliferation or differentiation. Together, our findings suggest that Cx43I130T/+ mice represent a surrogate model to not only inform about the craniofacial anomalies found in ODDD patients but also to show that reduced Cx43 function leads to phenotypic changes that are largely due to osteoblast defects.


Assuntos
Anormalidades Craniofaciais , Anormalidades Dentárias , Animais , Conexina 43/genética , Anormalidades Craniofaciais/genética , Anormalidades do Olho , Deformidades Congênitas do Pé , Junções Comunicantes , Humanos , Camundongos , Crânio , Sindactilia , Anormalidades Dentárias/genética
12.
Cell Rep ; 30(8): 2712-2728.e8, 2020 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-32101747

RESUMO

Histone deacetylases (HDACs) drive innate immune cell-mediated inflammation. Here we identify class IIa HDACs as key molecular links between Toll-like receptor (TLR)-inducible aerobic glycolysis and macrophage inflammatory responses. A proteomic screen identified the glycolytic enzyme pyruvate kinase M isoform 2 (Pkm2) as a partner of proinflammatory Hdac7 in murine macrophages. Myeloid-specific Hdac7 overexpression in transgenic mice amplifies lipopolysaccharide (LPS)-inducible lactate and promotes a glycolysis-associated inflammatory signature. Conversely, pharmacological or genetic targeting of Hdac7 and other class IIa HDACs attenuates LPS-inducible glycolysis and accompanying inflammatory responses in macrophages. We show that an Hdac7-Pkm2 complex acts as an immunometabolism signaling hub, whereby Pkm2 deacetylation at lysine 433 licenses its proinflammatory functions. Disrupting this complex suppresses inflammatory responses in vitro and in vivo. Class IIa HDACs are thus pivotal intermediates connecting TLR-inducible glycolysis to inflammation via Pkm2.


Assuntos
Glicólise , Histona Desacetilases/metabolismo , Inflamação/patologia , Macrófagos/enzimologia , Macrófagos/patologia , Piruvato Quinase/metabolismo , Receptores Toll-Like/metabolismo , Acetilação/efeitos dos fármacos , Animais , Glicólise/efeitos dos fármacos , Células HEK293 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/efeitos dos fármacos , Células RAW 264.7
13.
Biochem Cell Biol ; 87(4): 567-80, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19767821

RESUMO

The mouse F9 embryonal carcinoma cell line is ideally suited to study the epithelial-to-mesenchymal transition accompanying the differentiation of primitive to parietal extraembryonic endoderm. In F9 cells, the application of exogenous agents including retinoic acid or activation of signal transduction cascades downstream of G-proteins triggers widespread changes in gene expression and leads to the formation of primitive endoderm. The epithelial-to-mesenchymal transition is completed and parietal endoderm develops as of result of increasing PKA activity in primitive endoderm cells. Expression of a constitutively active form of Galpha13(Q226L) is sufficient to induce F9 cells into parietal endoderm and a model is emerging that a signaling axis linking G-protein signaling to RhoA and the ERM protein moesin is required for differentiation. In this study, we found that expression of either p115RhoGEF or a constitutively active, GTPase-deficient form of RhoA(L63) promoted primitive, but not parietal, endoderm formation. The overexpression of Galpha13(Q226L) or p115RhoGEF, but not Rho(L63), caused beta-catenin to translocate to the nucleus. Surprisingly, the stimulation of the Wnt-beta-catenin pathway was accompanied by nuclear beta-catenin and primitive endoderm formation, even when a dominant negative was used to block the signaling axis at the level of p115RhoGEF or when ROCK activity was inhibited using the pharmacological agent Y-27632. Together, results indicate that the coordinate signaling by two independent pathways, one involving canonical Wnt-beta-catenin activation of target genes and the other with Galpha13 signaling to ERM proteins to modulate cytoarchitectural changes, is required during the retinoic acid induced differentiation of F9 cells to primitive endoderm.


Assuntos
Diferenciação Celular , Endoderma/citologia , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Sequência de Bases , Linhagem Celular Tumoral , Imuno-Histoquímica , Camundongos , Dados de Sequência Molecular
14.
Cell Signal ; 20(3): 506-17, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18160257

RESUMO

Epithelial-to-mesenchymal transitions (EMTs) play key roles in the normal development of an organism as well as its demise following the metastasis of a malignant tumour. An EMT during early mouse development results in the differentiation of primitive endoderm into the parietal endoderm that forms part of the parietal yolk sac. In the embryo, primitive endoderm develops from cells in the inner cell mass, but the signals that instruct these cells to become specified and adopt an epithelial fate are poorly understood. The mouse F9 teratocarcinoma cell line, a model that can recapitulate the in vivo primitive to parietal endoderm EMT, has been used extensively to elucidate the signalling cascades involved in extraembryonic endoderm differentiation. Here, we identified Wnt6 as a gene up-regulated in F9 cells in response to RA and show that Wnt6 expressing cells or cells exposed to Wnt6 conditioned media form primitive endoderm. Wnt6 induction of primitive endoderm is accompanied by beta-catenin and Snail1 translocation to the nucleus and the appearance of cytokeratin intermediate filaments. Attenuating glycogen synthase kinase 3 activity using LiCl gave similar results, but the fact that cells de-differentiate when LiCl is removed reveals that other signalling pathways are required to maintain cells as primitive endoderm. Finally, Wnt6-induced primitive endodermal cells were tested to determine their competency to complete the EMT and differentiate into parietal endoderm. Towards that end, results show that up-regulating protein kinase A activity is sufficient to induce markers of parietal endoderm. Together, these findings indicate that undifferentiated F9 cells are responsive to canonical Wnt signalling, which negatively regulates glycogen synthase kinase 3 activity leading to the epithelialization and specification of primitive endoderm competent to receive additional signals required for EMT. Considering the ability of F9 cells to mimic an in vivo EMT, the identification of this Wnt6-beta-catenin-Snail signalling cascade has broad implications for understanding EMT mechanisms in embryogenesis and metastasis.


Assuntos
Padronização Corporal , Diferenciação Celular , Células-Tronco de Carcinoma Embrionário/metabolismo , Endoderma/metabolismo , Células Epiteliais/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Padronização Corporal/efeitos dos fármacos , Bucladesina/farmacologia , Células COS , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Chlorocebus aethiops , Meios de Cultivo Condicionados/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células-Tronco de Carcinoma Embrionário/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário/enzimologia , Células-Tronco de Carcinoma Embrionário/patologia , Endoderma/efeitos dos fármacos , Endoderma/enzimologia , Endoderma/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Regulação da Expressão Gênica no Desenvolvimento , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Filamentos Intermediários/metabolismo , Cloreto de Lítio/farmacologia , Camundongos , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo , Transfecção , Tretinoína/farmacologia , Regulação para Cima , Proteínas Wnt/genética , beta Catenina/metabolismo
15.
Cell Signal ; 20(1): 163-75, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17993262

RESUMO

The mouse F9 teratocarcinoma cell line is a model that can be manipulated to imitate one of the earliest epithelial-mesenchymal transitions in mouse development. When cells are treated with Retinoic Acid they differentiate into primitive endoderm and into parietal endoderm with the addition of dibutyryl cAMP. Parietal endoderm also develops when undifferentiated cells express a constitutively active (CA) form of Galpha13(Q226L). Differentiation is accompanied by a translocation of beta-catenin to the nucleus and considerable changes to the cytoskeleton and cell morphology. ERM proteins facilitate rearrangements to the F-actin cytoskeleton, and at least one, moesin, is essential for cell survival. In this study we found that moesin translocated to the nucleus during RA-induced differentiation, and sequence analysis identified putative nuclear localization signals in the protein. In the absence of RA, transient over-expression of rat moesin or the distantly related zebrafish homologue in F9 cells induced primitive endoderm. Furthermore, no apparent beta-catenin was seen in the nucleus of cells over-expressing zebrafish moesin. Our previous results have shown that depleting F9 cells of moesin using an antisense morpholino strategy caused them to detach from the substrate unless they expressed CA-Galpha13(Q226L). This CA-Galpha13 signalling maintained cell survival, but at the expense of differentiation. We now report that over-expressing zebrafish moesin in mouse moesin-depleted F9 cells not only ensured cell survival, but also induced differentiation to primitive endoderm. Together, the results suggest a new role for moesin, acting in a signalling pathway facilitating the differentiation of extraembryonic endoderm.


Assuntos
Diferenciação Celular/fisiologia , Endoderma/citologia , Proteínas dos Microfilamentos/metabolismo , Transdução de Sinais/fisiologia , Teratocarcinoma/patologia , Neoplasias Testiculares/patologia , Sequência de Aminoácidos , Animais , Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Endoderma/metabolismo , Masculino , Camundongos , Proteínas dos Microfilamentos/genética , Dados de Sequência Molecular , Teratocarcinoma/metabolismo , Neoplasias Testiculares/metabolismo , Transfecção , Tretinoína/farmacologia , Peixe-Zebra , beta Catenina/metabolismo
16.
Int J Dev Biol ; 52(8): 1143-50, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18956348

RESUMO

Heterotrimeric G-protein signaling, involving alpha, beta and gamma subunits, plays a number of roles in differentiation and development. Individual gamma subunits interact with a beta subunit and as a heterodimer, is responsible for modulating many G protein-mediated cellular responses. The 12 gamma subunits in mammals have highly variable distribution and expression patterns in adult tissues. gamma3 is abundantly and widely expressed in the brain and when its expression is knocked-out, the mice show increased susceptibility to seizures, reduced body weights and decreased adiposity compared to the wild-type littermates (Schwindinger et al., 2004). Recent evidence has shown the Gng3 gene being strongly induced in activated CD4+ T-cells (Dubeykovskiy et al., 2006) and its involvement in the developing mammalian enteric nervous system (Heanue and Pachnis, 2006). Given this diversity in expression and interest in finding models of human disease, and to extend our previous investigation with zebrafish gamma3 (Kelly et al., 2001), we undertook an analysis to report the temporal and spatial expression patterns of gamma3 mRNA during mouse embryogenesis. Analysis reveals that gamma3 transcripts were first expressed in mid-late embryonic stages. Specifically, signals were predominant in the CNS and in neural crest cell derivatives including but not limited to the trigeminal and dorsal root (spinal) ganglia, and in cells of the adrenal medulla. These data indicate that G protein coupled signaling involving gamma3 participates in a number of physiological roles, not only in the CNS, but also in numerous cells derived from the neural crest.


Assuntos
Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/genética , Crista Neural/embriologia , Crista Neural/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Sequência Conservada , Primers do DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Hibridização In Situ , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Crista Neural/citologia , Filogenia , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais
17.
Cell Death Discov ; 4: 42, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30302276

RESUMO

Glucose metabolism has a crucial role for providing substrates required to generate ATP and regulate the epigenetic landscape. We reported that F9 embryonal carcinoma stem-like cells require cytosolic reactive oxygen species to differentiate into extraembryonic endoderm; however, mitochondrial sources were not examined. To extend these studies, we examined the metabolic profile of early and late-passage F9 cells, and show that their ability to differentiate is similar, even though each population has dramatically different metabolic profiles. Differentiated early-passage cells relied on glycolysis, while differentiated late-passage cells transitioned towards oxidative phosphorylation (OXPHOS). Unexpectedly, electron transport chain protein stoichiometry was disrupted in differentiated late-passage cells, whereas genes encoding mitofusion 1 and 2, which promote mitochondrial fusion and favor OXPHOS, were upregulated in differentiated early-passage cells. Despite this, early-passage cells cultured under conditions to promote glycolysis showed enhanced differentiation, whereas promoting OXPHOS in late-passage cells showed a similar trend. Further analysis revealed that the distinct metabolic profiles seen between the two populations is largely associated with changes in genomic integrity, linking metabolism to passage number. Together, these results indicate that passaging has no effect on the potential for F9 cells to differentiate into extraembryonic endoderm; however, it does impact their metabolic profile. Thus, it is imperative to determine the molecular and metabolic status of a stem cell population before considering its utility as a therapeutic tool for regenerative medicine.

18.
Stem Cells Int ; 2017: 3684178, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28373885

RESUMO

Just over ten years have passed since the seminal Takahashi-Yamanaka paper, and while most attention nowadays is on induced, embryonic, and cancer stem cells, much of the pioneering work arose from studies with embryonal carcinoma cells (ECCs) derived from teratocarcinomas. This original work was broad in scope, but eventually led the way for us to focus on the components involved in the gene regulation of stemness and differentiation. As the name implies, ECCs are malignant in nature, yet maintain the ability to differentiate into the 3 germ layers and extraembryonic tissues, as well as behave normally when reintroduced into a healthy blastocyst. Retinoic acid signaling has been thoroughly interrogated in ECCs, especially in the F9 and P19 murine cell models, and while we have touched on this aspect, this review purposely highlights how some key transcription factors regulate pluripotency and cell stemness prior to this signaling. Another major focus is on the epigenetic regulation of ECCs and stem cells, and, towards that end, this review closes on what we see as a new frontier in combating aging and human disease, namely, how cellular metabolism shapes the epigenetic landscape and hence the pluripotency of all stem cells.

19.
PLoS One ; 12(2): e0170812, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28152080

RESUMO

Mouse F9 cells differentiate to primitive endoderm (PrE) when treated with retinoic acid (RA). Differentiation is accompanied by increased reactive oxygen species (ROS) levels, and while treating F9 cells with antioxidants attenuates differentiation, H2O2 treatment alone is sufficient to induce PrE. We identified the NADPH oxidase (NOX) complexes as candidates for the source of this endogenous ROS, and within this gene family, and over the course of differentiation, Nox1 and Nox 4 show the greatest upregulation induced by RA. Gata6, encoding a master regulator of extraembryonic endoderm is also up-regulated by RA and we provide evidence that NOX1 and NOX4 protein levels increase in F9 cells overexpressing Gata6. Pan-NOX and NOX1-specific inhibitors significantly reduced the ability of RA to induce PrE, and this was recapitulated using a genetic approach to knockdown Nox1 and/or Nox4 transcripts. Interestingly, overexpressing either gene in untreated F9 cells did not induce differentiation, even though each elevated ROS levels. Thus, the data suggests that ROS produced during PrE differentiation is dependent in part on increased NOX1 and NOX4 levels, which is under the control of GATA6. Furthermore, these results suggest that the combined activity of multiple NOX proteins is necessary for the differentiation of F9 cells to primitive endoderm.


Assuntos
Endoderma/citologia , Endoderma/metabolismo , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidases/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Endoderma/efeitos dos fármacos , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , NADH NADPH Oxirredutases/antagonistas & inibidores , NADH NADPH Oxirredutases/genética , NADPH Oxidase 1 , NADPH Oxidase 4 , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , Espécies Reativas de Oxigênio/metabolismo , Tretinoína/farmacologia , Via de Sinalização Wnt
20.
Front Cell Dev Biol ; 5: 93, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29119099

RESUMO

Mouse F9 cells differentiate into primitive extraembryonic endoderm (PrE) when treated with retinoic acid (RA), and this is accompanied by an up-regulation of Gata6. The role of the GATA6 network in PrE differentiation is known, and we have shown it directly activates Wnt6. Canonical Wnt/ß-catenin signaling is required by F9 cells to differentiate to PrE, and this, like most developmental processes, requires input from one or more additional pathways. We found both RA and Gata6 overexpression, can induce the expression of Indian Hedgehog (Ihh) and a subset of its target genes through Gli activation during PrE induction. Chemical activation of the Hh pathway using a Smoothened agonist (SAG) also increased Gli reporter activity, and as expected, when Hh signaling was blocked with a Smoothened antagonist, cyclopamine, this RA-induced reporter activity was reduced. Interestingly, SAG alone failed to induce markers of PrE differentiation, and had no effect on Wnt/ß-catenin-dependent TCF-LEF reporter activity. The expected increase in Wnt/ß-catenin-dependent TCF-LEF reporter activity and PrE markers induced by RA was, however, blocked by cyclopamine. Finally, inhibiting GSK3 activity with BIO increased both TCF-LEF and Gli reporter activities. Together, we demonstrate the involvement of Hh signaling in the RA-induced differentiation of F9 cells into PrE, and while the activation of the Hh pathway itself is not sufficient, it as well as active Wnt/ß-catenin are necessary for F9 cell differentiation.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA