Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Stem Cell Reports ; 18(11): 2240-2253, 2023 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-37922914

RESUMO

In early vertebrate development, organizer regions-groups of cells that signal to and thereby influence neighboring cells by secreted morphogens-play pivotal roles in the establishment and maintenance of cell identities within defined tissue territories. The midbrain-hindbrain organizer drives regionalization of neural tissue into midbrain and hindbrain territories with fibroblast growth factor 8 (FGF8) acting as a key morphogen. This organizer has been extensively studied in chicken, mouse, and zebrafish. Here, we demonstrate the enrichment of FGF8-expressing cells from human pluripotent stem cells (hPSCs), cultured as attached embryoid bodies using antibodies that recognize "Similar Expression to Fgf" (SEF) and Frizzled proteins. The arrangement of cells in embryoid body subsets of these cultures and the gene expression profile of the FGF8-expressing population show certain similarities to the midbrain-hindbrain organizer in animal models. In the embryonic chick brain, the enriched cell population induces formation of midbrain structures, consistent with FGF8-organizing capability.


Assuntos
Proteínas de Homeodomínio , Células-Tronco Pluripotentes , Humanos , Animais , Camundongos , Proteínas de Homeodomínio/metabolismo , Peixe-Zebra/metabolismo , Fator 8 de Crescimento de Fibroblasto/genética , Galinhas/metabolismo , Mesencéfalo/metabolismo , Células-Tronco Pluripotentes/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Crescimento de Fibroblastos/metabolismo , Padronização Corporal
2.
Front Cell Dev Biol ; 10: 941493, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36172281

RESUMO

Rett syndrome is a human intellectual disability disorder that is associated with mutations in the X-linked MECP2 gene. The epigenetic reader MeCP2 binds to methylated cytosines on the DNA and regulates chromatin organization. We have shown previously that MECP2 Rett syndrome missense mutations are impaired in chromatin binding and heterochromatin reorganization. Here, we performed a proteomics analysis of post-translational modifications of MeCP2 isolated from adult mouse brain. We show that MeCP2 carries various post-translational modifications, among them phosphorylation on S80 and S421, which lead to minor changes in either heterochromatin binding kinetics or clustering. We found that MeCP2 is (di)methylated on several arginines and that this modification alters heterochromatin organization. Interestingly, we identified the Rett syndrome mutation site R106 as a dimethylation site. In addition, co-expression of protein arginine methyltransferases (PRMT)1 and PRMT6 lead to a decrease of heterochromatin clustering. Altogether, we identified and validated novel modifications of MeCP2 in the brain and show that these can modulate its ability to bind as well as reorganize heterochromatin, which may play a role in the pathology of Rett syndrome.

3.
Nat Commun ; 10(1): 2966, 2019 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-31273213

RESUMO

Mutations in genes encoding components of BAF (BRG1/BRM-associated factor) chromatin remodeling complexes cause neurodevelopmental disorders and tumors. The mechanisms leading to the development of these two disease entities alone or in combination remain unclear. We generated mice with a heterozygous nervous system-specific partial loss-of-function mutation in a BAF core component gene, Smarcb1. These Smarcb1 mutant mice show various brain midline abnormalities that are also found in individuals with Coffin-Siris syndrome (CSS) caused by SMARCB1, SMARCE1, and ARID1B mutations and in SMARCB1-related intellectual disability (ID) with choroid plexus hyperplasia (CPH). Analyses of the Smarcb1 mutant animals indicate that one prominent midline abnormality, corpus callosum agenesis, is due to midline glia aberrations. Our results establish a novel role of Smarcb1 in the development of the brain midline and have important clinical implications for BAF complex-related ID/neurodevelopmental disorders.


Assuntos
Anormalidades Múltiplas/genética , Agenesia do Corpo Caloso/genética , Corpo Caloso/crescimento & desenvolvimento , Face/anormalidades , Deformidades Congênitas da Mão/genética , Deficiência Intelectual/genética , Micrognatismo/genética , Pescoço/anormalidades , Proteína SMARCB1/genética , Anormalidades Múltiplas/diagnóstico por imagem , Agenesia do Corpo Caloso/diagnóstico por imagem , Agenesia do Corpo Caloso/patologia , Alelos , Animais , Criança , Pré-Escolar , Corpo Caloso/citologia , Corpo Caloso/diagnóstico por imagem , Modelos Animais de Doenças , Embrião de Mamíferos , Face/diagnóstico por imagem , Feminino , Deformidades Congênitas da Mão/diagnóstico por imagem , Humanos , Lactente , Deficiência Intelectual/diagnóstico por imagem , Mutação com Perda de Função , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Transgênicos , Micrognatismo/diagnóstico por imagem , Pescoço/diagnóstico por imagem , Neuroglia/patologia , Cultura Primária de Células
4.
Nucleic Acids Res ; 45(12): 7118-7136, 2017 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-28449087

RESUMO

Cytosine modifications diversify and structure the genome thereby controlling proper development and differentiation. Here, we focus on the interplay of the 5-methylcytosine reader Mbd1 and modifier Tet1 by analyzing their dynamic subcellular localization and the formation of the Tet oxidation product 5-hydroxymethylcytosine in mammalian cells. Our results demonstrate that Mbd1 enhances Tet1-mediated 5-methylcytosine oxidation. We show that this is due to enhancing the localization of Tet1, but not of Tet2 and Tet3 at heterochromatic DNA. We find that the recruitment of Tet1 and concomitantly its catalytic activity eventually leads to the displacement of Mbd1 from methylated DNA. Finally, we demonstrate that increased Tet1 heterochromatin localization and 5-methylcytosine oxidation are dependent on the CXXC3 domain of Mbd1, which recognizes unmethylated CpG dinucleotides. The Mbd1 CXXC3 domain deletion isoform, which retains only binding to methylated CpGs, on the other hand, blocks Tet1-mediated 5-methylcytosine to 5-hydroxymethylcytosine conversion, indicating opposite biological effects of Mbd1 isoforms. Our study provides new insights on how cytosine modifications, their modifiers and readers cross-regulate themselves.


Assuntos
Ilhas de CpG , Proteínas de Ligação a DNA/genética , DNA/metabolismo , Regulação da Expressão Gênica , Heterocromatina/metabolismo , Oxigenases de Função Mista/genética , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição/genética , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animais , Linhagem Celular , DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/genética , Dioxigenases/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Heterocromatina/química , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Oxigenases de Função Mista/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Oxirredução , Domínios Proteicos , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteína Vermelha Fluorescente
5.
Oncotarget ; 7(36): 58203-58217, 2016 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-27533460

RESUMO

Bmi1 was originally identified as a gene that contributes to the development of mouse lymphoma by inhibiting MYC-induced apoptosis through repression of Ink4a and Arf. It codes for the Polycomb group protein BMI-1 and acts primarily as a transcriptional repressor via chromatin modifications. Although it binds to a large number of genomic regions, the direct BMI-1 target genes described so far do not explain the full spectrum of BMI-1-mediated effects. Here we identify the putative tumor suppressor gene EphA7 as a novel direct BMI-1 target in neural cells and lymphocytes. EphA7 silencing has been reported in several different human tumor types including lymphomas, and our data suggest BMI1 overexpression as a novel mechanism leading to EphA7 inactivation via H3K27 trimethylation and DNA methylation.


Assuntos
Regulação da Expressão Gênica , Genes Supressores de Tumor , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor EphA7/genética , Animais , Linfócitos B , Técnicas de Cultura de Células/métodos , Núcleo Celular/metabolismo , Proliferação de Células/fisiologia , Células Cultivadas , Cerebelo/anatomia & histologia , Cerebelo/metabolismo , Metilação de DNA/fisiologia , Regulação para Baixo , Histonas/metabolismo , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Ventrículos Laterais/anatomia & histologia , Ventrículos Laterais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Células-Tronco Neurais , Complexo Repressor Polycomb 1/genética , Proteínas Proto-Oncogênicas/genética , Receptor EphA7/metabolismo , Baço/citologia , Transdução Genética , Regulação para Cima
6.
Biochem Biophys Res Commun ; 478(2): 573-9, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27470587

RESUMO

Podocalyxin (PODXL) is a highly glycosylated and sialylated transmembrane protein that is up-regulated in various types of tumors and whose expression levels positively correlate with tumor grade. We previously found Podxl to be highly expressed in murine tumorigenic neural stem/progenitor cells (NSPs). Here we investigated the effects of elevated Podxl levels in these cells. NSPs overexpressing Podxl did not form brain tumors upon intracranial transplantations, indicating that high levels of this gene alone are not sufficient for tumor initiation. However, Podxl overexpression had a positive effect on cell number, sphere formation and cell viability, indicating that it might in this way contribute to the development and/or maintenance of tumors. Proteome analyses of Podxl-overexpressing and control NSPs revealed increased levels of Annexin A2 (ANXA2). We also found increased transcript levels, indicating that PODXL stimulates expression of the Anxa2 gene. Lack of Anxa2 in Podxl-overexpressing NSPs resulted in reduced viability of these cells, suggesting that PODXL-mediated pro-survival effects can at least in part be explained by increased ANXA2 levels. Finally, our data indicate that Podxl overexpression activates the MAP kinase (MAPK) pathway which in turn up-regulates Anxa2 expression. Our data indicate a novel molecular connection between PODXL and ANXA2: both exert pro-survival effects in NSPs, and PODXL positively regulates ANXA2 expression through the MAPK pathway.


Assuntos
Anexina A2/genética , Sobrevivência Celular , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neurais/metabolismo , Sialoglicoproteínas/genética , Regulação para Cima , Animais , Anexina A2/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular , Proliferação de Células , Células Cultivadas , Regulação para Baixo , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/patologia , Sialoglicoproteínas/metabolismo , Ativação Transcricional
7.
Stem Cells Dev ; 23(16): 1844-57, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24809660

RESUMO

True tendon regeneration in human patients remains a vision of musculoskeletal therapies. In comparison to other mesenchymal lineages the biology of tenogenic differentiation is barely understood. Specifically, easy and efficient protocols are lacking that might enable tendon cell and tissue differentiation based on adult (stem) cell sources. In the murine mesenchymal progenitor cell line C3H10T½, overexpression of the growth factor bone morphogenetic protein 2 (BMP2) and a constitutively active transcription factor, Smad8 L+MH2, mediates tendon cell differentiation in vitro and the formation of tendon-like tissue in vivo. We hypothesized that during this differentiation secreted factors involved in extracellular matrix formation exert a major impact on tendon development. Gene expression analyses revealed four genes encoding secreted factors that are notably upregulated: periostin, C-type lectin domain family 3 (member b), RNase A4, and follistatin-like 1. These factors have not previously been implicated in tendon biology. Among these, periostin showed a specific expression in tenocytes of adult mouse Achilles tendon and in chondrocytes within the nonmineralized fibrocartilage zone of the enthesis with the calcaneus. Overexpression of periostin alone or in combination with constitutively active BMP receptor type in human mesenchymal stem cells and subsequent implantation into ectopic sites in mice demonstrated a reproducible moderate tenogenic capacity that has not been described before. Therefore, periostin may belong to the factors contributing to the development of tenogenic tissue.


Assuntos
Tendão do Calcâneo/fisiopatologia , Moléculas de Adesão Celular/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Animais , Osso e Ossos/metabolismo , Moléculas de Adesão Celular/fisiologia , Diferenciação Celular , Endorribonucleases/genética , Endorribonucleases/metabolismo , Feminino , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Expressão Gênica , Células HEK293 , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Camundongos Nus , Regeneração , Regulação para Cima
8.
Diabetes ; 62(8): 2834-42, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23610061

RESUMO

Precise regulation of ß-cell function is crucial for maintaining blood glucose homeostasis. Pax6 is an essential regulator of ß-cell-specific factors like insulin and Glut2. Studies in the developing eye suggest that Pax6 interacts with Mitf to regulate pigment cell differentiation. Here, we show that Mitf, like Pax6, is expressed in all pancreatic endocrine cells during mouse postnatal development and in the adult islet. A Mitf loss-of-function mutation results in improved glucose tolerance and enhanced insulin secretion but no increase in ß-cell mass in adult mice. Mutant ß-cells secrete more insulin in response to glucose than wild-type cells, suggesting that Mitf is involved in regulating ß-cell function. In fact, the transcription of genes critical for maintaining glucose homeostasis (insulin and Glut2) and ß-cell formation and function (Pax4 and Pax6) is significantly upregulated in Mitf mutant islets. The increased Pax6 expression may cause the improved ß-cell function observed in Mitf mutant animals, as it activates insulin and Glut2 transcription. Chromatin immunoprecipitation analysis shows that Mitf binds to Pax4 and Pax6 regulatory regions, suggesting that Mitf represses their transcription in wild-type ß-cells. We demonstrate that Mitf directly regulates Pax6 transcription and controls ß-cell function.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Fator de Transcrição Associado à Microftalmia/metabolismo , Mutação , Animais , Glicemia/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Camundongos , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Ativação Transcricional
9.
Nat Med ; 18(8): 1232-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22820645

RESUMO

Primary astrocytomas of grade 3 or 4 according to the classification system of the World Health Organization (high-grade astrocytomas or HGAs) are preponderant among adults and are almost invariably fatal despite the use of multimodal therapy. Here we show that the juvenile brain has an endogenous defense mechanism against HGAs. Neural precursor cells (NPCs) migrate to HGAs, reduce glioma expansion and prolong survival time by releasing endovanilloids that activate the vanilloid receptor (transient receptor potential vanilloid subfamily member-1 or TRPV1) on HGA cells. TRPV1 is highly expressed in tumor and weakly expressed in tumor-free brain. TRPV1 stimulation triggers tumor cell death through the branch of the endoplasmic reticulum stress pathway that is controlled by activating transcription factor-3 (ATF3). The antitumorigenic response of NPCs is lost with aging. NPC-mediated tumor suppression can be mimicked in the adult brain by systemic administration of the synthetic vanilloid arvanil, suggesting that TRPV1 agonists have potential as new HGA therapeutics.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Proteínas de Neoplasias/fisiologia , Células-Tronco Neurais/fisiologia , Canais de Cátion TRPV/fisiologia , Envelhecimento/metabolismo , Amidas , Amidoidrolases/deficiência , Amidoidrolases/genética , Animais , Antineoplásicos/uso terapêutico , Apoptose , Ácidos Araquidônicos/metabolismo , Ácidos Araquidônicos/farmacologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Capsaicina/uso terapêutico , Movimento Celular , Meios de Cultivo Condicionados/farmacologia , Dopamina/análogos & derivados , Dopamina/metabolismo , Dopamina/farmacologia , Endocanabinoides/metabolismo , Endocanabinoides/farmacologia , Etanolaminas/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Células-Tronco Neurais/metabolismo , Ácidos Oleicos/metabolismo , Ácidos Oleicos/farmacologia , Ácidos Palmíticos/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/análise , Canais de Cátion TRPV/biossíntese , Canais de Cátion TRPV/genética , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia
10.
Cancer Res ; 72(13): 3381-92, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22719073

RESUMO

Although brain tumors are classified and treated based upon their histology, the molecular factors involved in the development of various tumor types remain unknown. In this study, we show that the type and order of genetic events directs the development of gliomas, central nervous system primitive neuroectodermal tumors, and atypical teratoid/rhabdoid-like tumors from postnatal mouse neural stem/progenitor cells (NSC/NPC). We found that the overexpression of specific genes led to the development of these three different brain tumors from NSC/NPCs, and manipulation of the order of genetic events was able to convert one established tumor type into another. In addition, loss of the nuclear chromatin-remodeling factor SMARCB1 in rhabdoid tumors led to increased phosphorylation of eIF2α, a central cytoplasmic unfolded protein response (UPR) component, suggesting a role for the UPR in these tumors. Consistent with this, application of the proteasome inhibitor bortezomib led to an increase in apoptosis of human cells with reduced SMARCB1 levels. Taken together, our findings indicate that the order of genetic events determines the phenotypes of brain tumors derived from a common precursor cell pool, and suggest that the UPR may represent a therapeutic target in atypical teratoid/rhabdoid tumors.


Assuntos
Neoplasias Encefálicas/patologia , Células-Tronco Neurais/citologia , Animais , Linhagem da Célula , Montagem e Desmontagem da Cromatina , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
J Neurosci ; 32(15): 5151-64, 2012 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-22496561

RESUMO

Ischemic stroke causes transient increase of neural stem and progenitor cell (NSPC) proliferation in the subventricular zone (SVZ), and migration of newly formed neuroblasts toward the damaged area where they mature to striatal neurons. The molecular mechanisms regulating this plastic response, probably involved in structural reorganization and functional recovery, are poorly understood. The adaptor protein LNK suppresses hematopoietic stem cell self-renewal, but its presence and role in the brain are poorly understood. Here we demonstrate that LNK is expressed in NSPCs in the adult mouse and human SVZ. Lnk(-/-) mice exhibited increased NSPC proliferation after stroke, but not in intact brain or following status epilepticus. Deletion of Lnk caused increased NSPC proliferation while overexpression decreased mitotic activity of these cells in vitro. We found that Lnk expression after stroke increased in SVZ through the transcription factors STAT1/3. LNK attenuated insulin-like growth factor 1 signaling by inhibition of AKT phosphorylation, resulting in reduced NSPC proliferation. Our findings identify LNK as a stroke-specific, endogenous negative regulator of NSPC proliferation, and suggest that LNK signaling is a novel mechanism influencing plastic responses in postischemic brain.


Assuntos
Isquemia Encefálica/patologia , Encéfalo/citologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Células-Tronco Neurais/fisiologia , Acidente Vascular Cerebral/patologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antimetabólitos , Bromodesoxiuridina , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Eletroporação , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/patologia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Knockout , Proteína Oncogênica v-akt/genética , Proteína Oncogênica v-akt/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Recuperação de Função Fisiológica , Retroviridae/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/fisiologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/fisiologia , Fatores de Transcrição/metabolismo , Transfecção/métodos
12.
Hum Mol Genet ; 21(8): 1673-80, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22186023

RESUMO

Rett syndrome (RTT) is caused by loss-of-function mutations in the X-linked gene MECP2 coding for methyl CpG-binding protein 2 (MeCP2). This protein can act as transcriptional repressor, and we showed in a previous study that glucocorticoid-inducible genes are up-regulated in an RTT mouse model and that these genes are direct MeCP2 targets. Here, we report that pharmacological intervention with the glucocorticoid system has an impact on the symptoms and lifespan in an RTT mouse model. Our data support a functional implication of the stress hormone system in RTT and suggest this hormone system as potential therapeutic target.


Assuntos
Corticosterona/farmacologia , Glucocorticoides/metabolismo , Mifepristona/farmacologia , Receptores de Glucocorticoides/metabolismo , Síndrome de Rett/fisiopatologia , Animais , Corticosterona/administração & dosagem , Corticosterona/sangue , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Proteínas Imediatamente Precoces/genética , Expectativa de Vida , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Mifepristona/metabolismo , Atividade Motora/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Receptores de Glucocorticoides/antagonistas & inibidores , Síndrome de Rett/genética , Teste de Desempenho do Rota-Rod , Proteínas de Ligação a Tacrolimo/genética
13.
Glia ; 59(1): 68-81, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21046556

RESUMO

In contrast to ependymal cells located above the subventricular zone (SVZ) of the adult lateral ventricle wall (LVW), adult spinal cord (SC) ependymal cells possess certain neural stem cell characteristics. The molecular basis of this difference is unknown. In this study, antibodies against multiple cell surface markers were applied to isolate pure populations of SC and LVW ependymal cells, which allowed a direct comparison of their in vitro behavior and in vivo gene expression profile. Isolated CD133(+)/CD24(+)/CD45(-)/CD34(-) ependymal cells from the SC displayed in vitro self-renewal and differentiation capacity, whereas those from the LVW did not. SC ependymal cells showed a higher expression of several genes involved in cell division, cell cycle regulation, and chromosome stability, which is consistent with a long-term self-renewal capacity, and shared certain transcripts with neural stem cells of the embryonic forebrain. They also expressed several retinoic acid (RA)-regulated genes and responded to RA exposure. LVW ependymal cells showed higher transcript levels of many genes regulated by transforming growth factor-ß family members. Among them were Dlx2, Id2, Hey1, which together with Foxg1 could explain their potential to turn into neuroblasts under certain environmental conditions.


Assuntos
Antígenos CD/metabolismo , Antígeno CD24/metabolismo , Epêndima/citologia , Expressão Gênica , Glicoproteínas/metabolismo , Ventrículos Laterais/citologia , Peptídeos/metabolismo , Medula Espinal/citologia , Antígeno AC133 , Animais , Diferenciação Celular , Células Cultivadas , Epêndima/metabolismo , Citometria de Fluxo , Imuno-Histoquímica , Ventrículos Laterais/metabolismo , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/metabolismo
14.
Nucleic Acids Res ; 37(11): e82, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19443447

RESUMO

The prediction of transcription factor binding sites in genomic sequences is in principle very useful to identify upstream regulatory factors. However, when applying this concept to genomes of multicellular organisms such as mammals, one has to deal with a large number of false positive predictions since many transcription factor genes are only expressed in specific tissues or cell types. We developed TS-REX, a database/software system that supports the analysis of tissue and cell type-specific transcription factor-gene networks based on expressed sequence tag abundance of transcription factor-encoding genes in UniGene EST libraries. The use of expression levels of transcription factor-encoding genes according to hierarchical anatomical classifications covering different tissues and cell types makes it possible to filter out irrelevant binding site predictions and to identify candidates of potential functional importance for further experimental testing. TS-REX covers ESTs from H. sapiens and M. musculus, and allows the characterization of both presence and specificity of transcription factors in user-specified tissues or cell types. The software allows users to interactively visualize transcription factor-gene networks, as well as to export data for further processing. TS-REX was applied to predict regulators of Polycomb group genes in six human tumor tissues and in human embryonic stem cells.


Assuntos
Bases de Dados Genéticas , Redes Reguladoras de Genes , Software , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Células-Tronco Embrionárias/metabolismo , Etiquetas de Sequências Expressas , Regulação da Expressão Gênica , Biblioteca Gênica , Humanos , Camundongos , Neoplasias/genética , Neoplasias/metabolismo , Proteínas do Grupo Polycomb , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição/genética
15.
Cancer Res ; 67(12): 5727-36, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17575139

RESUMO

Human brain tumor stem cells have been enriched using antibodies against the surface protein CD133. An antibody recognizing CD133 also served to isolate normal neural stem cells from fetal human brain, suggesting a possible lineage relationship between normal neural and brain tumor stem cells. Whether CD133-positive brain tumor stem cells can be derived from CD133-positive neural stem or progenitor cells still requires direct experimental evidence, and an important step toward such investigations is the identification and characterization of normal CD133-presenting cells in neurogenic regions of the embryonic and adult brain. Here, we present evidence that CD133 is a marker for embryonic neural stem cells, an intermediate radial glial/ependymal cell type in the early postnatal stage, and for ependymal cells in the adult brain, but not for neurogenic astrocytes in the adult subventricular zone. Our findings suggest two principal possibilities for the origin of brain tumor stem cells: a derivation from CD133-expressing cells, which are normally not present in the adult brain (embryonic neural stem cells and an early postnatal intermediate radial glial/ependymal cell type), or from CD133-positive ependymal cells in the adult brain, which are, however, generally regarded as postmitotic. Alternatively, brain tumor stem cells could be derived from proliferative but CD133-negative neurogenic astrocytes in the adult brain. In the latter case, brain tumor development would involve the production of CD133.


Assuntos
Antígenos CD/metabolismo , Neoplasias Encefálicas/metabolismo , Células-Tronco Embrionárias/metabolismo , Epêndima/metabolismo , Glioblastoma/metabolismo , Glicoproteínas/metabolismo , Peptídeos/metabolismo , Prosencéfalo/metabolismo , Antígeno AC133 , Adulto , Animais , Astrócitos/metabolismo , Western Blotting , Epêndima/citologia , Células Epiteliais/metabolismo , Citometria de Fluxo , Imunofluorescência , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neoplásicas/metabolismo , Neuroglia/metabolismo , Prosencéfalo/citologia , Prosencéfalo/crescimento & desenvolvimento
16.
PLoS Comput Biol ; 2(9): e123, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16978048

RESUMO

Recent ChIP experiments of human and mouse embryonic stem cells have elucidated the architecture of the transcriptional regulatory circuitry responsible for cell determination, which involves the transcription factors OCT4, SOX2, and NANOG. In addition to regulating each other through feedback loops, these genes also regulate downstream target genes involved in the maintenance and differentiation of embryonic stem cells. A search for the OCT4-SOX2-NANOG network motif in other species reveals that it is unique to mammals. With a kinetic modeling approach, we ascribe function to the observed OCT4-SOX2-NANOG network by making plausible assumptions about the interactions between the transcription factors at the gene promoter binding sites and RNA polymerase (RNAP), at each of the three genes as well as at the target genes. We identify a bistable switch in the network, which arises due to several positive feedback loops, and is switched on/off by input environmental signals. The switch stabilizes the expression levels of the three genes, and through their regulatory roles on the downstream target genes, leads to a binary decision: when OCT4, SOX2, and NANOG are expressed and the switch is on, the self-renewal genes are on and the differentiation genes are off. The opposite holds when the switch is off. The model is extremely robust to parameter changes. In addition to providing a self-consistent picture of the transcriptional circuit, the model generates several predictions. Increasing the binding strength of NANOG to OCT4 and SOX2, or increasing its basal transcriptional rate, leads to an irreversible bistable switch: the switch remains on even when the activating signal is removed. Hence, the stem cell can be manipulated to be self-renewing without the requirement of input signals. We also suggest tests that could discriminate between a variety of feedforward regulation architectures of the target genes by OCT4, SOX2, and NANOG.


Assuntos
Células-Tronco Embrionárias/metabolismo , Transcrição Gênica/genética , Animais , Biologia Computacional , Simulação por Computador , Redes Reguladoras de Genes , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Homeodomínio/genética , Humanos , Modelos Biológicos , Fator 3 de Transcrição de Octâmero/genética , Filogenia
17.
Dev Dyn ; 234(4): 1034-45, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16247769

RESUMO

Different causes, such as maternal diabetes, cloning by nuclear transfer, interspecific hybridization, and deletion of some genes such as Esx1, Ipl, or Cdkn1c, may underlie placental overgrowth. In a previous study, we carried out comparative gene expression analysis in three models of placental hyperplasias, cloning, interspecies hybridization (IHPD), and Esx1 deletion. This study identified a large number of genes that exhibited differential expression between normal and enlarged placentas; however, it remained unclear how altered expression of any specific gene was related to any specific placental phenotype. In the present study, we focused on two genes, Car2 and Ncam1, which both exhibited increased expression in interspecies and cloned hyperplastic placentas. Apart from a detailed expression analysis of both genes during normal murine placentation, we also assessed morphology of placentas that were null for Car2 or Ncam1. Finally, we attempted to rescue placental hyperplasia in a congenic model of IHPD by decreasing transcript levels of Car2 or Ncam1. In situ analysis showed that both genes are expressed mainly in the spongiotrophoblast, however, expression patterns exhibited significant variability during development. Contrary to expectations, homozygous deletion of either Car2 or Ncam1 did not result in placental phenotypes. However, expression analysis of Car3 and Ncam2, which can take over the function of Car2 and Ncam1, respectively, indicated a possible rescue mechanism, as Car3 and Ncam2 were expressed in spongiotrophoblast of Car2 and Ncam1 mutant placentas. On the other hand, downregulation of either Car2 or Ncam1 did not rescue any of the placental phenotypes of AT24 placentas, a congenic model for interspecies hybrid placentas. This strongly suggested that altered expression of Car2 and Ncam1 is a downstream event in placental hyperplasia.


Assuntos
Antígeno CD56/metabolismo , Anidrase Carbônica II/metabolismo , Regulação da Expressão Gênica , Fenótipo , Doenças Placentárias/genética , Animais , Antígeno CD56/genética , Anidrase Carbônica II/genética , Primers do DNA , Feminino , Hibridização In Situ , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Doenças Placentárias/patologia , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA
18.
Hum Mol Genet ; 14(15): 2247-56, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16002417

RESUMO

Rett syndrome (RTT) is a severe form of mental retardation, which is caused by spontaneous mutations in the X-linked gene MECP2. How the loss of MeCP2 function leads to RTT is currently unknown. Mice lacking the Mecp2 gene initially show normal postnatal development but later acquire neurological phenotypes, including heightened anxiety, that resemble RTT. The MECP2 gene encodes a methyl-CpG-binding protein that can act as a transcriptional repressor. Using cDNA microarrays, we found that Mecp2-null animals differentially express several genes that are induced during the stress response by glucocorticoids. Increased levels of mRNAs for serum glucocorticoid-inducible kinase 1 (Sgk) and FK506-binding protein 51 (Fkbp5) were observed before and after onset of neurological symptoms, but plasma glucocorticoid was not significantly elevated in Mecp2-null mice. MeCP2 is bound to the Fkbp5 and Sgk genes in brain and may function as a modulator of glucocorticoid-inducible gene expression. Given the known deleterious effect of glucocorticoid exposure on brain development, our data raise the possibility that disruption of MeCP2-dependent regulation of stress-responsive genes contributes to the symptoms of RTT.


Assuntos
Corticosterona/sangue , Regulação da Expressão Gênica , Proteínas Imediatamente Precoces/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Síndrome de Rett/metabolismo , Estresse Fisiológico/sangue , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Encéfalo/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Síndrome de Rett/genética , Transdução de Sinais , Regulação para Cima
19.
Stem Cells ; 23(6): 842-51, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15917480

RESUMO

We recently established that two midgestation-derived stromal clones--UG26-1B6, urogenital ridge-derived, and EL08-1D2, embryonic liver-derived--support the maintenance of murine adult bone marrow and human cord blood hematopoietic repopulating stem cells (HSCs). In this study, we investigate whether direct HSC-stroma contact is required for this stem cell maintenance. Adult bone marrow ckit+ Ly-6C- side population (K6-SP) cells and stromal cells were cocultured under contact or noncontact conditions. These experiments showed that HSCs were maintained for at least 4 weeks in culture and that direct contact between HSCs and stromal cells was not required. To find out which factors might be involved in HSC maintenance, we compared the gene expression profile of EL08-1D2 and UG26-1B6 with four HSC-nonsupportive clones. We found that EL08-1D2 and UG26-1B6 both expressed 21 genes at a higher level, including the putative secreted factors fibroblast growth factor-7, insulin-like growth factor-binding proteins 3 and 4, pleiotrophin, pentaxin-related, and thrombospondin 2, whereas 11 genes, including GPX-3 and HSP27, were expressed at a lower level. In summary, we show for the first time long-term maintenance of adult bone marrow HSCs in stroma noncontact cultures and identify some secreted molecules that may be involved in this support.


Assuntos
Embrião de Mamíferos/citologia , Células-Tronco Hematopoéticas/citologia , Células Estromais/citologia , Animais , Células da Medula Óssea/citologia , Proteína C-Reativa/metabolismo , Proteínas de Transporte/metabolismo , Técnicas de Cultura de Células/métodos , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Primers do DNA/química , Ensaio de Imunoadsorção Enzimática , Sangue Fetal/citologia , Fator 7 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/metabolismo , Glutationa Peroxidase/metabolismo , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/metabolismo , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Fígado/citologia , Fígado/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Chaperonas Moleculares , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Ligação Proteica , RNA/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia , Trombospondinas/metabolismo , Fatores de Tempo
20.
Eur J Cell Biol ; 84(2-3): 123-35, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15819395

RESUMO

Epigenetics is a term that has changed its meaning with the increasing biological knowledge on developmental processes. However, its current application to stem cell biology is often imprecise and is conceptually problematic. This article addresses two different subjects, the definition of epigenetics and chromatin states of stem and differentiated cells. We describe mechanisms that regulate chromatin changes and provide an overview of chromatin states of stem and differentiated cells. Moreover, a modification of the current epigenetics definition is proposed that is not restricted by the heritability of gene expression throughout cell divisions and excludes translational gene expression control.


Assuntos
Cromatina/fisiologia , Epigênese Genética/fisiologia , Células-Tronco/fisiologia , Animais , Cromatina/genética , Montagem e Desmontagem da Cromatina/fisiologia , Proteínas Cromossômicas não Histona/fisiologia , Metilação de DNA , Regulação da Expressão Gênica/fisiologia , Histonas/fisiologia , Humanos , RNA não Traduzido/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA