Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Life Sci Alliance ; 5(12)2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-36104080

RESUMO

Progeroid syndromes are rare genetic diseases with most of autosomal dominant transmission, the prevalence of which is less than 1/10,000,000. These syndromes caused by mutations in the <i>LMNA</i> gene encoding A-type lamins belong to a group of disorders called laminopathies. Lamins are implicated in the architecture and function of the nucleus and chromatin. Patients affected with progeroid laminopathies display accelerated aging of mesenchymal stem cells (MSCs)-derived tissues associated with nuclear morphological abnormalities. To identify pathways altered in progeroid patients' MSCs, we used induced pluripotent stem cells (hiPSCs) from patients affected with classical Hutchinson-Gilford progeria syndrome (HGPS, c.1824C&gt;T-p.G608G), HGPS-like syndrome (HGPS-L; c.1868C&gt;G-p.T623S) associated with farnesylated prelamin A accumulation, or atypical progeroid syndromes (APS; homozygous c.1583C&gt; T-p.T528M; heterozygous c.1762T&gt;C-p.C588R; compound heterozygous c.1583C&gt;T and c.1619T&gt;C-p.T528M and p.M540T) without progerin accumulation. By comparative analysis of the transcriptome and methylome of hiPSC-derived MSCs, we found that patient's MSCs display specific DNA methylation patterns and modulated transcription at early stages of differentiation. We further explored selected biological processes deregulated in the presence of <i>LMNA</i> variants and confirmed alterations of age-related pathways during MSC differentiation. In particular, we report the presence of an altered mitochondrial pattern; an increased response to double-strand DNA damage; and telomere erosion in HGPS, HGPS-L, and APS MSCs, suggesting converging pathways, independent of progerin accumulation, but a distinct DNA methylation profile in HGPS and HGPS-L compared with APS cells.


Assuntos
Senilidade Prematura , Células-Tronco Mesenquimais , Progéria , Envelhecimento/genética , Senilidade Prematura/genética , Humanos , Células-Tronco Mesenquimais/metabolismo , Progéria/metabolismo , Síndrome
2.
Cell Death Dis ; 12(7): 649, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34172716

RESUMO

Endothelial-mesenchymal transition (EndMT) is an important source of cancer-associated fibroblasts (CAFs), which facilitates tumour progression. PDAC is characterised by abundant CAFs and tumour necrosis factor-α (TNF-α). Here, we show that TNF-α strongly induces human endothelial cells to undergo EndMT. Interestingly, TNF-α strongly downregulates the expression of the endothelial receptor TIE1, and reciprocally TIE1 overexpression partially prevents TNF-α-induced EndMT, suggesting that TNF-α acts, at least partially, through TIE1 regulation in this process. We also show that TNF-α-induced EndMT is reversible. Furthermore, TNF-α treatment of orthotopic mice resulted in an important increase in the stroma, including CAFs. Finally, secretome analysis identified TNFSF12, as a regulator that is also present in PDAC patients. With the aim of restoring normal angiogenesis and better access to drugs, our results support the development of therapies targeting CAFs or inducing the EndMT reversion process in PDAC.


Assuntos
Fibroblastos Associados a Câncer/efeitos dos fármacos , Carcinoma Ductal Pancreático/patologia , Células Endoteliais/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias Pancreáticas/patologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Células Cultivadas , Citocina TWEAK/genética , Citocina TWEAK/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Masculino , Camundongos Transgênicos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Receptor de TIE-1/genética , Receptor de TIE-1/metabolismo , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Homeobox 2 de Ligação a E-box com Dedos de Zinco/genética , Homeobox 2 de Ligação a E-box com Dedos de Zinco/metabolismo
3.
Nucleic Acids Res ; 47(6): 2822-2839, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30698748

RESUMO

The DNA methylation epigenetic signature is a key determinant during development. Rules governing its establishment and maintenance remain elusive especially at repetitive sequences, which account for the majority of methylated CGs. DNA methylation is altered in a number of diseases including those linked to mutations in factors that modify chromatin. Among them, SMCHD1 (Structural Maintenance of Chromosomes Hinge Domain Containing 1) has been of major interest following identification of germline mutations in Facio-Scapulo-Humeral Dystrophy (FSHD) and in an unrelated developmental disorder, Bosma Arhinia Microphthalmia Syndrome (BAMS). By investigating why germline SMCHD1 mutations lead to these two different diseases, we uncovered a role for this factor in de novo methylation at the pluripotent stage. SMCHD1 is required for the dynamic methylation of the D4Z4 macrosatellite upon reprogramming but seems dispensable for methylation maintenance. We find that FSHD and BAMS patient's cells carrying SMCHD1 mutations are both permissive for DUX4 expression, a transcription factor whose regulation has been proposed as the main trigger for FSHD. These findings open new questions as to what is the true aetiology for FSHD, the epigenetic events associated with the disease thus calling the current model into question and opening new perspectives for understanding repetitive DNA sequences regulation.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Metilação de DNA , Proteínas de Homeodomínio/genética , Repetições de Microssatélites/genética , Células Cultivadas , Reprogramação Celular/genética , Atresia das Cóanas/genética , Atresia das Cóanas/metabolismo , Metilação de DNA/genética , Epigênese Genética/genética , Regulação da Expressão Gênica , Células HCT116 , Células HEK293 , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Microftalmia/genética , Microftalmia/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/metabolismo , Distrofia Muscular Facioescapuloumeral/patologia , Nariz/anormalidades
4.
Sci Rep ; 6: 32490, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27577850

RESUMO

Human induced pluripotent stem cells (hiPSCs) show great promise for obesity treatment as they represent an unlimited source of brown/brite adipose progenitors (BAPs). However, hiPSC-BAPs display a low adipogenic capacity compared to adult-BAPs when maintained in a traditional adipogenic cocktail. The reasons of this feature are unknown and hamper their use both in cell-based therapy and basic research. Here we show that treatment with TGFß pathway inhibitor SB431542 together with ascorbic acid and EGF were required to promote hiPSCs-BAP differentiation at a level similar to adult-BAP differentiation. hiPSC-BAPs expressed the molecular identity of adult-UCP1 expressing cells (PAX3, CIDEA, DIO2) with both brown (ZIC1) and brite (CD137) adipocyte markers. Altogether, these data highlighted the critical role of TGFß pathway in switching off hiPSC-brown adipogenesis and revealed novel factors to unlock their differentiation. As hiPSC-BAPs display similarities with adult-BAPs, it opens new opportunities to develop alternative strategies to counteract obesity.


Assuntos
Adipócitos Marrons/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/antagonistas & inibidores , Adipócitos Marrons/citologia , Adipócitos Marrons/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Ácido Ascórbico/farmacologia , Benzamidas/farmacologia , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Dioxóis/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Iodeto Peroxidase/genética , Iodeto Peroxidase/metabolismo , Fator de Transcrição PAX3/genética , Fator de Transcrição PAX3/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Iodotironina Desiodinase Tipo II
5.
Stem Cells Transl Med ; 3(12): 1467-72, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25355730

RESUMO

For years, our ability to study pathological changes in neurological diseases has been hampered by the lack of relevant models until the recent groundbreaking work from Yamanaka's group showing that it is feasible to generate induced pluripotent stem cells (iPSCs) from human somatic cells and to redirect the fate of these iPSCs into differentiated cells. In particular, much interest has focused on the ability to differentiate human iPSCs into neuronal progenitors and functional neurons for relevance to a large number of pathologies including mental retardation and behavioral or degenerative syndromes. Current differentiation protocols are time-consuming and generate limited amounts of cells, hindering use on a large scale. We describe a feeder-free method relying on the use of a chemically defined medium that overcomes the need for embryoid body formation and neuronal rosette isolation for neuronal precursors and terminally differentiated neuron production. Four days after induction, expression of markers of the neurectoderm lineage is detectable. Between 4 and 7 days, neuronal precursors can be expanded, frozen, and thawed without loss of proliferation and differentiation capacities or further differentiated. Terminal differentiation into the different subtypes of mature neurons found in the human brain were observed. At 6-35 days after induction, cells express typical voltage-gated and ionotrophic receptors for GABA, glycine, and acetylcholine. This specific and efficient single-step strategy in a chemically defined medium allows the production of mature neurons in 20-40 days with multiple applications, especially for modeling human pathologies.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Técnicas de Cultura de Células , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/citologia , Neurônios/metabolismo , Fatores de Tempo
6.
PLoS One ; 7(11): e49065, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23145071

RESUMO

Regulation of the extracellular matrix (ECM) plays an important functional role either in physiological or pathological conditions. The plasminogen activation (PA) system, comprising the uPA and tPA proteases and their inhibitor PAI-1, is one of the main suppliers of extracellular proteolytic activity contributing to tissue remodeling. Although its function in development is well documented, its precise role in mouse embryonic stem cell (ESC) differentiation in vitro is unknown. We found that the PA system components are expressed at very low levels in undifferentiated ESCs and that upon differentiation uPA activity is detected mainly transiently, whereas tPA activity and PAI-1 protein are maximum in well differentiated cells. Adipocyte formation by ESCs is inhibited by amiloride treatment, a specific uPA inhibitor. Likewise, ESCs expressing ectopic PAI-1 under the control of an inducible expression system display reduced adipogenic capacities after induction of the gene. Furthermore, the adipogenic differentiation capacities of PAI-1(-/-) induced pluripotent stem cells (iPSCs) are augmented as compared to wt iPSCs. Our results demonstrate that the control of ESC adipogenesis by the PA system correspond to different successive steps from undifferentiated to well differentiated ESCs. Similarly, skeletal myogenesis is decreased by uPA inhibition or PAI-1 overexpression during the terminal step of differentiation. However, interfering with uPA during days 0 to 3 of the differentiation process augments ESC myotube formation. Neither neurogenesis, cardiomyogenesis, endothelial cell nor smooth muscle formation are affected by amiloride or PAI-1 induction. Our results show that the PA system is capable to specifically modulate adipogenesis and skeletal myogenesis of ESCs by successive different molecular mechanisms.


Assuntos
Adipogenia/fisiologia , Células-Tronco Embrionárias/fisiologia , Desenvolvimento Muscular/fisiologia , Ativadores de Plasminogênio/genética , Ativadores de Plasminogênio/metabolismo , Plasminogênio/genética , Plasminogênio/metabolismo , Adipócitos/metabolismo , Animais , Diferenciação Celular/genética , Células-Tronco Embrionárias/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Serpina E2/genética , Serpina E2/metabolismo
7.
EMBO Rep ; 13(5): 431-9, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22421998

RESUMO

Endothelial-mesenchymal transition (EndMT) has a significant role in embryonic heart formation and in various pathologies. However, the molecular mechanisms that regulate EndMT induction remain to be elucidated. We show that suppression of receptor tyrosine kinase Tie1 but not Tie2 induces human endothelial cells to undergo EndMT and that Slug deficiency reverts this process. We find that Erk1/2, Erk5 and Akt cascades control Slug promoter activity induced by Tie1 deficiency. Interestingly, EndMT is present in human pancreatic tumour. We propose that EndMT associated with Tie1 downregulation participates in the pathological development of stroma observed in tumours.


Assuntos
Transdiferenciação Celular/fisiologia , Endotélio/patologia , Mesoderma/patologia , Neovascularização Patológica/metabolismo , Receptor de TIE-1/metabolismo , Adesão Celular/genética , Adesão Celular/fisiologia , Movimento Celular/genética , Movimento Celular/fisiologia , Transdiferenciação Celular/genética , Células Cultivadas , Endotélio/metabolismo , Humanos , Immunoblotting , Masculino , Mesoderma/metabolismo , Microscopia Confocal , Neovascularização Patológica/genética , Regiões Promotoras Genéticas/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Receptor de TIE-1/genética , Transdução de Sinais , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Stem Cells Dev ; 20(7): 1233-46, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20954847

RESUMO

Embryonic stem (ES) cells differentiate in vitro into all cell lineages. We previously found that the p38 mitogen activated kinase (p38MAPK) pathway controls the commitment of ES cells toward either cardiomyogenesis (p38 on) or neurogenesis (p38 off ). In this study, we show that p38α knock-out ES cells do not differentiate into cardiac, endothelial, smooth muscle, and skeletal muscle lineages. Reexpression of p38MAPK in these cells partially rescues their mesodermal differentiation defects and corrects the high level of spontaneous neurogenesis of knock-out cells. Wild-type ES cells were treated with a p38MAPK-specific inhibitor during the differentiation process. These experiments allowed us to identify 2 early independent successive p38MAPK functions in the formation of mesodermal lineages. Further, the first one correlates with the regulation of the expression of Brachyury, an essential mesodermal-specific transcription factor, by p38MAPK. In conclusion, by genetic and biochemical approaches, we demonstrate that p38MAPK activity is essential for the commitment of ES cell into cardiac, endothelial, smooth muscle, and skeletal muscle mesodermal lineages.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Mesoderma/citologia , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Animais , Western Blotting , Células Cultivadas , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Citometria de Fluxo , Imunofluorescência , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Imidazóis/farmacologia , Mesoderma/metabolismo , Camundongos , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Desenvolvimento Muscular , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
9.
Stem Cells Cloning ; 3: 103-4, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-24198515
10.
Stem Cells ; 25(5): 1090-5, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17218395

RESUMO

Embryonic stem (ES) cells can give rise, in vivo, to the ectodermal, endodermal, and mesodermal germ layers and, in vitro, can differentiate into multiple cell lineages, offering broad perspectives in regenerative medicine. Understanding the molecular mechanisms governing ES cell commitment is an essential challenge in this field. The mitogen-activated protein kinase (MAPK) pathways extracellular signal-regulated kinase (ERK), c-Jun amino-terminal kinase (JNK), and p38MAPK are able to regulate ES commitment from early steps of the process to mature differentiated cells. Whereas the ERK pathway inhibits the self-renewal of ES cells, upon commitment this pathway is involved in the development of extraembryonic tissues, in early mesoderm differentiation, and in the formation of mature adipocytes; p38MAPK displays a large spectrum of action from neurons to adipocytes, and JNK is involved in both ectoderm and primitive endoderm differentiations. Furthermore, for a given pathway, several of these effects are isoform-dependent, revealing the complexity of the cellular response to activation of MAPK pathways. Regarding tissue regeneration, the potential outcome of systematic analysis of the function of different MAPKs in different ES cell differentiation programs is discussed. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Linhagem da Célula , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Sistema de Sinalização das MAP Quinases , Modelos Biológicos
11.
J Soc Biol ; 200(1): 93-7, 2006.
Artigo em Francês | MEDLINE | ID: mdl-17144167

RESUMO

Obesity is often associated with diabetes and insulin resistance. This review summarizes evidence obtained in our lab on the role of the serine phosphorylation of the insulin receptor substrate 1 in the down regulation of insulin signalling. The role of the ERK1 isoform in the development of adipose tissue and insulin sensitivity is also presented.


Assuntos
Diabetes Mellitus/fisiopatologia , Resistência à Insulina/fisiologia , Insulina/fisiologia , Obesidade/fisiopatologia , Transdução de Sinais/fisiologia , Tecido Adiposo/fisiologia , Tecido Adiposo/fisiopatologia , Humanos , Proteínas Substratos do Receptor de Insulina , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfoproteínas/fisiologia
12.
Antimicrob Agents Chemother ; 50(12): 3998-4004, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17000748

RESUMO

Highly active antiretroviral therapy (HAART) of human immunodeficiency virus-infected patients is associated with adverse effects, such as lipodystrophy and hyperlipidemia. The lipodystrophic syndrome is characterized by a peripheral lipoatrophy and/or fat accumulation in the abdomen and neck. In order to get insights into the physiopathological mechanisms underlying this syndrome, we treated mice with protease inhibitors (PIs) over a long period of time. Although atazanavir-treated mice presented the same circulating triglyceride concentration as control mice, lopinavir-ritonavir-treated mice rapidly became hypertriglyceridemic, with triglyceride levels of 200 mg/dl, whereas control and atazanavir-treated animals had triglyceride levels of 80 mg/dl. These results obtained with mice reproduce the metabolic disorder observed in humans. White adipose tissue (WAT) was analyzed after 8 weeks of treatment. Compared to the control or atazanavir treatment, lopinavir-ritonavir treatment induced a significant 25% weight reduction in the peripheral inguinal WAT depot. By contrast, the profound epididymal WAT depot was not affected. This effect was associated with a 5.5-fold increase in SREBP-1c gene expression only in the inguinal depot. Our results demonstrate that the long-term treatment of mice with PIs constitutes an interesting experimental model with which some aspects of the lipoatrophy induced by HAART in humans may be studied.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Inibidores da Protease de HIV/farmacologia , Pirimidinonas/farmacologia , Ritonavir/farmacologia , Animais , Sulfato de Atazanavir , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores da Protease de HIV/sangue , Inibidores da Protease de HIV/farmacocinética , Hipertrigliceridemia/tratamento farmacológico , Hipertrigliceridemia/etiologia , Lopinavir , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oligopeptídeos/sangue , Oligopeptídeos/farmacocinética , Oligopeptídeos/farmacologia , Piridinas/sangue , Piridinas/farmacocinética , Piridinas/farmacologia , Pirimidinonas/sangue , Pirimidinonas/farmacocinética , RNA Mensageiro/análise , Ritonavir/sangue , Ritonavir/farmacocinética , Fatores de Tempo , Triglicerídeos/sangue
13.
Diabetes ; 55(2): 281-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16443758

RESUMO

Formation of new adipocytes from precursor cells contributes to adipose tissue expansion and obesity. In this study, we asked whether p38 mitogen-activated protein kinase (MAPK) pathway regulates normal and pathological adipogenesis. In both dietary and genetically (ob/ob) obese mice, adipose tissues displayed a marked decrease in p38MAPK activity compared with the same tissues from lean mice. Furthermore, p38MAPK activity was significantly higher in preadipocytes than in adipocytes, suggesting that p38MAPK activity decreases during adipocyte differentiation. In agreement with an inhibitory role of p38MAPK in this process, we found that in vitro inhibition of p38MAPK, with the specific inhibitor PD169316, increased the expression of adipocyte markers in several cellular models, from embryonic to adult stages. Importantly, the expression of adipocyte markers was higher in p38MAPKalpha knockout cells than in their wild-type counterparts. Phosphorylation of C/EBPbeta, which enhances its transcriptional activity, is increased after p38MAPK inhibition. Finally, either inhibition or disruption of p38MAPK increased peroxisome proliferator-activated receptor (PPAR)gamma expression and transactivation. Rescue of p38MAPK in knockout cells reduced PPARgamma activity to the low basal level of wild-type cells. We demonstrate here, by using multipronged approaches involving p38 chemical inhibitor and p38MAPKalpha knockout cells, that p38MAPK plays a negative role in adipogenesis via inhibition of C/EBPbeta and PPARgamma transcriptional activities.


Assuntos
Adipogenia/fisiologia , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Células 3T3-L1 , Adipócitos/enzimologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Diferenciação Celular , Fibroblastos/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Camundongos , Proteína Quinase 14 Ativada por Mitógeno/genética , PPAR gama/metabolismo , Células-Tronco/metabolismo
14.
Stem Cells ; 24(5): 1399-406, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16424397

RESUMO

Mouse embryonic stem (ES) cells can be differentiated, in vitro into a variety of cell types including cardiac cells and neurons. This process is strictly controlled by the potent morphogen retinoic acid (RA). At a concentration of 10(-7) M, RA induces ES cell differentiation into neurons and, conversely, inhibits cardiomyogenesis. We found that p38 mitogen-activated protein kinase (p38MAPK) activity peaked spontaneously, between day 3 and day 5, during ES cell differentiation and that RA completely inhibited this peak of activity. In contrast to wild-type cells, which required RA treatment, p38alpha(-/-) ES cells differentiated spontaneously into neurons and did not form cardiomyocytes. Moreover, inhibition of the peak of p38MAPK activity by a specific inhibitor, PD169316, committed ES cells into the neuronal lineage and blocked cardiomyogenesis. By genetic and biochemical approaches, we demonstrate that, in two different ES cell lines, the control of p38MAPK activity constitutes an early switch, committing ES cells into either neurogenesis (p38 off) or cardiomyogenesis (p38 on).


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/citologia , Neurônios/citologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/efeitos dos fármacos , Camundongos , Camundongos Knockout , Tretinoína/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética
15.
Oncogene ; 24(41): 6281-91, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16007198

RESUMO

The high mobility group type A-2 (HMGA 2) transcription factor is involved in proliferation and differentiation, mainly during embryogenesis. Its activated form (HMGA 2/T) presents oncogenic activities both in vivo and in vitro. However, its precise role during embryogenesis is unknown. We investigated its role during the commitment of mouse embryonic stem (ES) cells by constructing cell lines expressing either wild type (wt) or HMGA 2/T forms of the gene. Following differentiation, control and wt HMGA 2 ES cells did not display myotubes; whereas HMGA 2/T ES cell lines massively formed contractile myotubes. Furthermore, as opposed to control cells, HMGA 2/T ES cells highly expressed the muscle myosin heavy chain (MHC) marker. Interestingly, in experimental conditions inhibitory for myogenesis, we observed a strong expression of MyoD and myogenin in HMGA 2/T cells. By contrast, commitment into adipocyte, neuron, and cardiomyocyte lineages was not affected. Teratocarcinomas induced by HMGA 2/T ES cell lines presented numerous skeletal muscle-differentiated tissues that were not observed in wt HMGA 2 or control tumours. Finally, rapamycin, an inhibitor of the mTOR kinase, downregulated endogenous HMGA-2 expression and inhibited myogenesis. This effect was prevented by overexpression of exogenous HMGA-2. Our results reveal a novel function of HMGA-2 in skeletal muscle differentiation.


Assuntos
Diferenciação Celular/fisiologia , Embrião de Mamíferos/citologia , Proteína HMGA2/fisiologia , Fibras Musculares Esqueléticas/citologia , Oncogenes , Células-Tronco/citologia , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Camundongos , Teratocarcinoma/patologia
16.
Diabetes ; 54(2): 402-11, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15677498

RESUMO

Hyperplasia of adipose tissue is critical for the development of obesity, but molecular mechanisms governing normal or pathological recruitment of new adipocytes remain unclear. The extracellular signal-regulated kinase (ERK) pathway plays a pivotal role in many essential cellular functions, such as proliferation and differentiation. Using ERK1(-/-) mice, we investigated the role of this isoform in adipose tissue development. Mice lacking ERK1 have decreased adiposity and fewer adipocytes than wild-type animals. Furthermore, ERK1(-/-) mice challenged with high-fat diet are resistant to obesity, are protected from insulin resistance, and have a higher postprandial metabolic rate. To get insights into cellular mechanisms implicated in reduced adiposity in ERK1(-/-) animals, we analyzed adipocyte differentiation in ERK1(-/-) cells. Compared with wild-type control cells, mouse embryo fibroblasts and cultures of adult preadipocytes isolated from ERK1(-/-) adult animals exhibit impaired adipogenesis. An inhibitor of the ERK pathway does not affect the residual adipogenesis of the ERK1(-/-) cells, suggesting that ERK2 is not implicated in adipocyte differentiation. Our results clearly link ERK1 to the regulation of adipocyte differentiation, adiposity, and high-fat diet-induced obesity. This suggests that a therapeutic approach of obesity targeting specifically the ERK1 isoform and not ERK2 would be of particular interest.


Assuntos
Adipócitos/fisiologia , Tecido Adiposo/fisiologia , Gorduras na Dieta/farmacologia , Metabolismo Energético/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Adipócitos/citologia , Tecido Adiposo/embriologia , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Diferenciação Celular , Cruzamentos Genéticos , Embrião de Mamíferos , Teste de Tolerância a Glucose , Insulina/farmacologia , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 3 Ativada por Mitógeno/deficiência , Proteína Quinase 3 Ativada por Mitógeno/genética , Atividade Motora , Células-Tronco/citologia , Células-Tronco/fisiologia
17.
Biochem J ; 361(Pt 3): 621-7, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11802792

RESUMO

Mouse embryonic stem (ES) cells are pluripotent cells that differentiate into multiple cell lineages. The commitment of ES cells into the adipocyte lineage is dependent on an early 3-day treatment with all-trans retinoic acid (RA). To characterize the molecular mechanisms underlying this process, we examined the contribution of the extracellular-signal-regulated kinase (ERK) pathway. Treatment of ES cell-derived embryoid bodies with RA resulted in a prolonged activation of the ERK pathway, but not the c-Jun N-terminal kinase, p38 mitogen-activated protein kinase or phosphoinositide 3-kinase pathways. To investigate the role of ERK activation, co-treatment of RA with PD98059, a specific inhibitor of the ERK signalling pathway, prevented both adipocyte formation and expression of the adipogenic markers, adipocyte lipid-binding protein and peroxisome-proliferator-activated receptor gamma. Furthermore, we show that ERK activation is required only during RA treatment. PD98059 does not interfere with the commitment of ES cells into other lineages, such as neurogenesis, myogenesis and cardiomyogenesis. As opposed to the controversial role of the ERK pathway in terminal differentiation, our results clearly demonstrate that this pathway is specifically required at an early stage of adipogenesis, corresponding to the RA-dependent commitment of ES cells.


Assuntos
Adipócitos/citologia , Embrião de Mamíferos/citologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células-Tronco/citologia , Tretinoína/metabolismo , Adipócitos/metabolismo , Animais , Butadienos/farmacologia , Linhagem da Célula , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Imidazóis/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Miocárdio/citologia , Neurônios/metabolismo , Nitrilas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica , Piridinas/farmacologia , RNA/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição/metabolismo , Tretinoína/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...