Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Development ; 145(1)2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29158447

RESUMO

Although cardiac neural crest cells are required at early stages of arterial valve development, their contribution during valvular leaflet maturation remains poorly understood. Here, we show in mouse that neural crest cells from pre-otic and post-otic regions make distinct contributions to the arterial valve leaflets. Genetic fate-mapping analysis of Krox20-expressing neural crest cells shows a large contribution to the borders and the interleaflet triangles of the arterial valves. Loss of Krox20 function results in hyperplastic aortic valve and partially penetrant bicuspid aortic valve formation. Similar defects are observed in neural crest Krox20-deficient embryos. Genetic lineage tracing in Krox20-/- mutant mice shows that endothelial-derived cells are normal, whereas neural crest-derived cells are abnormally increased in number and misplaced in the valve leaflets. In contrast, genetic ablation of Krox20-expressing cells is not sufficient to cause an aortic valve defect, suggesting that adjacent cells can compensate this depletion. Our findings demonstrate a crucial role for Krox20 in arterial valve development and reveal that an excess of neural crest cells may be associated with bicuspid aortic valve.


Assuntos
Valva Aórtica/anormalidades , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Células Endoteliais/metabolismo , Doenças das Valvas Cardíacas/embriologia , Miocárdio/metabolismo , Crista Neural/metabolismo , Animais , Valva Aórtica/citologia , Valva Aórtica/embriologia , Doença da Válvula Aórtica Bicúspide , Proteína 2 de Resposta de Crescimento Precoce/genética , Células Endoteliais/citologia , Camundongos , Camundongos Knockout , Miocárdio/citologia , Crista Neural/citologia
2.
PLoS Genet ; 14(8): e1007581, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30080860

RESUMO

Cis-regulation plays an essential role in the control of gene expression, and is particularly complex and poorly understood for developmental genes, which are subject to multiple levels of modulation. In this study, we performed a global analysis of the cis-acting elements involved in the control of the zebrafish developmental gene krox20. krox20 encodes a transcription factor required for hindbrain segmentation and patterning, a morphogenetic process highly conserved during vertebrate evolution. Chromatin accessibility analysis reveals a cis-regulatory landscape that includes 6 elements participating in the control of initiation and autoregulatory aspects of krox20 hindbrain expression. Combining transgenic reporter analyses and CRISPR/Cas9-mediated mutagenesis, we assign precise functions to each of these 6 elements and provide a comprehensive view of krox20 cis-regulation. Three important features emerged. First, cooperation between multiple cis-elements plays a major role in the regulation. Cooperation can surprisingly combine synergy and redundancy, and is not restricted to transcriptional enhancer activity (for example, 4 distinct elements cooperate through different modes to maintain autoregulation). Second, several elements are unexpectedly versatile, which allows them to be involved in different aspects of control of gene expression. Third, comparative analysis of the elements and their activities in several vertebrate species reveals that this versatility is underlain by major plasticity across evolution, despite the high conservation of the gene expression pattern. These characteristics are likely to be of broad significance for developmental genes.


Assuntos
Proteína 2 de Resposta de Crescimento Precoce/genética , Regulação da Expressão Gênica no Desenvolvimento , Rombencéfalo/metabolismo , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Sistemas CRISPR-Cas , Cromatina/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/fisiologia , Elementos Facilitadores Genéticos , Evolução Molecular , Loci Gênicos , Morfogênese/genética , Ativação Transcricional , Peixe-Zebra/embriologia
3.
Int J Mol Sci ; 22(23)2021 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-34884531

RESUMO

Central nervous system (CNS) lesions are a leading cause of death and disability worldwide. Three-dimensional neural cultures in biomaterials offer more physiologically relevant models for disease studies, toxicity screenings or in vivo transplantations. Herein, we describe the development and use of pullulan/dextran polysaccharide-based scaffolds for 3D neuronal culture. We first assessed scaffolding properties upon variation of the concentration (1%, 1.5%, 3% w/w) of the cross-linking agent, sodium trimetaphosphate (STMP). The lower STMP concentration (1%) allowed us to generate scaffolds with higher porosity (59.9 ± 4.6%), faster degradation rate (5.11 ± 0.14 mg/min) and lower elastic modulus (384 ± 26 Pa) compared with 3% STMP scaffolds (47 ± 2.1%, 1.39 ± 0.03 mg/min, 916 ± 44 Pa, respectively). Using primary cultures of embryonic neurons from PGKCre, Rosa26tdTomato embryos, we observed that in 3D culture, embryonic neurons remained in aggregates within the scaffolds and did not attach, spread or differentiate. To enhance neuronal adhesion and neurite outgrowth, we then functionalized the 1% STMP scaffolds with laminin. We found that treatment of the scaffold with a 100 µg/mL solution of laminin, combined with a subsequent freeze-drying step, created a laminin mesh network that significantly enhanced embryonic neuron adhesion, neurite outgrowth and survival. Such scaffold therefore constitutes a promising neuron-compatible and biodegradable biomaterial.


Assuntos
Materiais Biocompatíveis/química , Técnicas de Cultura de Células em Três Dimensões/métodos , Embrião de Mamíferos/citologia , Neurônios/citologia , Polissacarídeos/química , Alicerces Teciduais/química , Animais , Adesão Celular , Sobrevivência Celular , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Porosidade , Engenharia Tecidual
4.
PLoS Genet ; 13(7): e1006903, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28749941

RESUMO

Developmental genes can harbour multiple transcriptional enhancers that act simultaneously or in succession to achieve robust and precise spatiotemporal expression. However, the mechanisms underlying cooperation between cis-acting elements are poorly documented, notably in vertebrates. The mouse gene Krox20 encodes a transcription factor required for the specification of two segments (rhombomeres) of the developing hindbrain. In rhombomere 3, Krox20 is subject to direct positive feedback governed by an autoregulatory enhancer, element A. In contrast, a second enhancer, element C, distant by 70 kb, is active from the initiation of transcription independent of the presence of the KROX20 protein. Here, using both enhancer knock-outs and investigations of chromatin organisation, we show that element C possesses a dual activity: besides its classical enhancer function, it is also permanently required in cis to potentiate the autoregulatory activity of element A, by increasing its chromatin accessibility. This work uncovers a novel, asymmetrical, long-range mode of cooperation between cis-acting elements that might be essential to avoid promiscuous activation of positive autoregulatory elements.


Assuntos
Proteína 1 de Resposta de Crescimento Precoce/genética , Elementos Facilitadores Genéticos , Elementos Reguladores de Transcrição/genética , Rombencéfalo/crescimento & desenvolvimento , Animais , Padronização Corporal/genética , Cromatina/genética , Proteína 1 de Resposta de Crescimento Precoce/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Camundongos Knockout , Mutação , Rombencéfalo/metabolismo , Homologia de Sequência do Ácido Nucleico
5.
Acta Neuropathol ; 138(3): 457-476, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31011859

RESUMO

Schwann cells (SC) enter the central nervous system (CNS) in pathophysiological conditions. However, how SC invade the CNS to remyelinate central axons remains undetermined. We studied SC migratory behavior ex vivo and in vivo after exogenous transplantation in the demyelinated spinal cord. The data highlight for the first time that SC migrate preferentially along blood vessels in perivascular extracellular matrix (ECM), avoiding CNS myelin. We demonstrate in vitro and in vivo that this migration route occurs by virtue of a dual mode of action of Eph/ephrin signaling. Indeed, EphrinB3, enriched in myelin, interacts with SC Eph receptors, to drive SC away from CNS myelin, and triggers their preferential adhesion to ECM components, such as fibronectin via integrinß1 interactions. This complex interplay enhances SC migration along the blood vessel network and together with lesion-induced vascular remodeling facilitates their timely invasion of the lesion site. These novel findings elucidate the mechanism by which SC invade and contribute to spinal cord repair.


Assuntos
Vasos Sanguíneos , Movimento Celular/fisiologia , Efrina-B3/metabolismo , Remielinização/fisiologia , Células de Schwann/fisiologia , Medula Espinal/metabolismo , Animais , Doenças Desmielinizantes/patologia , Feminino , Fibronectinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais/fisiologia , Medula Espinal/patologia
6.
Development ; 142(1): 185-95, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25516974

RESUMO

Although many components of the genetic pathways that provide positional information during embryogenesis have been identified, it remains unclear how these signals are integrated to specify discrete tissue territories. Here, we investigate the molecular mechanisms underlying the formation of one of the hindbrain segments, rhombomere (r) 3, specified by the expression of the gene krox20. Dissecting krox20 transcriptional regulation has identified several input pathways: Hox paralogous 1 (PG1) factors, which both directly activate krox20 and indirectly repress it via Nlz factors, and the molecular components of an Fgf-dependent effector pathway. These different inputs are channelled through a single initiator enhancer element to shape krox20 initial transcriptional response: Hox PG1 and Nlz factors define the anterior-posterior extent of the enhancer's domain of activity, whereas Fgf signalling modulates the magnitude of activity in a spatially uniform manner. Final positioning of r3 boundaries requires interpretation of this initial pattern by a krox20 positive-feedback loop, orchestrated by another enhancer. Overall, this study shows how positional information provided by different patterning mechanisms is integrated through a gene regulatory network involving two cis-acting elements operating on the same gene, thus offering a comprehensive view of the delimitation of a territory.


Assuntos
Padronização Corporal/genética , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento , Modelos Biológicos , Transdução de Sinais/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
7.
Eur J Immunol ; 43(4): 1063-73, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23349024

RESUMO

Interleukin-27 (IL-27) suppresses immune responses through inhibition of the development of IL-17 producing Th17 cells and induction of IL-10 production. We previously showed that forced expression of early growth response gene 2 (Egr-2), a transcription factor required for T-cell anergy induction, induces IL-10 and lymphocyte activation gene 3 expression and confers regulatory activity on CD4(+) T cells in vivo. Here, we evaluated the role of Egr-2 in IL-27-induced IL-10 production. Among various IL-10-inducing factors, only IL-27 induced high levels of Egr-2 and lymphocyte activation gene 3 expression. Intriguingly, IL-27 failed to induce IL-10 in Egr-2-deficient T cells. IL-27-mediated induction of Prdm1 that codes B lymphocyte induced maturation protein-1, a transcriptional regulator important for IL-10 production in CD4(+) T cells, was also impaired in the absence of Egr-2. Although IL-27-mediated IL-10 induction was dependent on both STAT1 and STAT3, only STAT3 was required for IL-27-mediated Egr-2 induction. These results suggest that IL-27 signal transduction through Egr-2 and B lymphocyte induced maturation protein-1 plays an important role in IL-10 production. Furthermore, Egr-2-deficient CD4(+) T cells showed dysregulated production of IFN-γ and IL-17 in response to IL-27 stimulation. Therefore, Egr-2 may play key roles in controlling the balance between regulatory and effector cytokines.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Interleucina-10/biossíntese , Interleucina-17/farmacologia , Fatores de Transcrição/metabolismo , Animais , Antígenos CD/metabolismo , Linfócitos T CD4-Positivos/imunologia , Proteína 2 de Resposta de Crescimento Precoce/genética , Interferon gama/biossíntese , Interleucina-17/biossíntese , Camundongos , Camundongos Knockout , Fator 1 de Ligação ao Domínio I Regulador Positivo , Regiões Promotoras Genéticas , Ligação Proteica , Fator de Transcrição STAT3/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional , Proteína do Gene 3 de Ativação de Linfócitos
8.
Development ; 138(2): 317-26, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21177344

RESUMO

Vertebrate hindbrain segmentation is an evolutionarily conserved process that involves a complex interplay of transcription factors and signalling pathways. Fibroblast growth factor (FGF) signalling plays a major role, notably by controlling the expression of the transcription factor Krox20 (Egr2), which is required for the formation and specification of two segmental units: rhombomeres (r) 3 and 5. Here, we explore the molecular mechanisms downstream of FGF signalling and the function of Sprouty 4 (Spry4), a negative-feedback regulator of this pathway, in zebrafish. We show that precise modulation of FGF signalling by Spry4 is required to determine the appropriate onset of krox20 transcription in r3 and r5 and, ultimately, rhombomere size in the r3-r5 region. FGF signalling acts by modulating the activity of krox20 initiator enhancer elements B and C; in r5, we show that this regulation is mediated by direct binding of the transcription factor MafB to element B. By contrast, FGF signalling does not control the krox20 autoregulatory element A, which is responsible for amplification and maintenance of krox20 expression. Therefore, early krox20 transcription sets the blueprint for r3-r5 patterning. This work illustrates the necessity for fine-tuning in a common and fundamental patterning process, based on a bistable cell-fate choice involving the coupling of an extracellular gradient with a positive-feedback loop. In this mode of patterning, precision and robustness can be achieved by the introduction of a negative-feedback loop, which, in the hindbrain, is mediated by Spry4.


Assuntos
Proteína 2 de Resposta de Crescimento Precoce/genética , Proteínas do Tecido Nervoso/metabolismo , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Sequência de Bases , Sítios de Ligação/genética , Padronização Corporal/genética , Padronização Corporal/fisiologia , Embrião de Galinha , Primers do DNA/genética , Elementos Facilitadores Genéticos , Retroalimentação Fisiológica , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fator de Transcrição MafB/genética , Fator de Transcrição MafB/metabolismo , Família Multigênica , Proteínas do Tecido Nervoso/genética , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Transcrição Gênica , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
9.
Mol Syst Biol ; 9: 690, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24061538

RESUMO

Although feedback loops are essential in development, their molecular implementation and precise functions remain elusive. Using enhancer knockout in mice, we demonstrate that a direct, positive autoregulatory loop amplifies and maintains the expression of Krox20, a transcription factor governing vertebrate hindbrain segmentation. By combining quantitative data collected in the zebrafish with biophysical modelling that accounts for the intrinsic stochastic molecular dynamics, we dissect the loop at the molecular level. We find that it underpins a bistable switch that turns a transient input signal into cell fate commitment, as we observe in single cell analyses. The stochasticity of the activation process leads to a graded input-output response until saturation is reached. Consequently, the duration and strength of the input signal controls the size of the hindbrain segments by modulating the distribution between the two cell fates. Moreover, segment formation is buffered from severe variations in input level. Finally, the progressive extinction of Krox20 expression involves a destabilization of the loop by repressor molecules. These mechanisms are of general significance for cell type specification and tissue patterning.


Assuntos
Padronização Corporal/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 2 de Resposta de Crescimento Precoce/genética , Retroalimentação Fisiológica , Regulação da Expressão Gênica no Desenvolvimento , Rombencéfalo/citologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Proliferação de Células , Embrião de Galinha , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Embrião de Mamíferos , Embrião não Mamífero , Elementos Facilitadores Genéticos , Hibridização In Situ , Camundongos , Dados de Sequência Molecular , Rombencéfalo/crescimento & desenvolvimento , Rombencéfalo/metabolismo , Transdução de Sinais , Processos Estocásticos , Transcrição Gênica , Peixe-Zebra
10.
Nature ; 451(7177): 460-4, 2008 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-18185516

RESUMO

Stem cell self-renewal implies proliferation under continued maintenance of multipotency. Small changes in numbers of stem cells may lead to large differences in differentiated cell numbers, resulting in significant physiological consequences. Proliferation is typically regulated in the G1 phase, which is associated with differentiation and cell cycle arrest. However, embryonic stem (ES) cells may lack a G1 checkpoint. Regulation of proliferation in the 'DNA damage' S/G2 cell cycle checkpoint pathway is known for its role in the maintenance of chromatin structural integrity. Here we show that autocrine/paracrine gamma-aminobutyric acid (GABA) signalling by means of GABA(A) receptors negatively controls ES cell and peripheral neural crest stem (NCS) cell proliferation, preimplantation embryonic growth and proliferation in the boundary-cap stem cell niche, resulting in an attenuation of neuronal progenies from this stem cell niche. Activation of GABA(A) receptors leads to hyperpolarization, increased cell volume and accumulation of stem cells in S phase, thereby causing a rapid decrease in cell proliferation. GABA(A) receptors signal through S-phase checkpoint kinases of the phosphatidylinositol-3-OH kinase-related kinase family and the histone variant H2AX. This signalling pathway critically regulates proliferation independently of differentiation, apoptosis and overt damage to DNA. These results indicate the presence of a fundamentally different mechanism of proliferation control in these stem cells, in comparison with most somatic cells, involving proteins in the DNA damage checkpoint pathway.


Assuntos
Histonas/metabolismo , Receptores de GABA-A/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Comunicação Autócrina , Blastocisto/citologia , Blastocisto/enzimologia , Blastocisto/metabolismo , Contagem de Células , Ciclo Celular , Linhagem Celular , Proliferação de Células , Tamanho Celular , Dano ao DNA , Agonistas de Receptores de GABA-A , Antagonistas de Receptores de GABA-A , Histonas/deficiência , Histonas/genética , Camundongos , Crista Neural/citologia , Crista Neural/metabolismo , Comunicação Parácrina , Técnicas de Patch-Clamp , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Receptores de GABA-A/genética , Células-Tronco/enzimologia , Ácido gama-Aminobutírico/metabolismo
11.
Proc Natl Acad Sci U S A ; 108(26): 10714-9, 2011 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-21670295

RESUMO

Boundary cap cells (BC), which express the transcription factor Krox20, participate in the formation of the boundary between the central nervous system and the peripheral nervous system. To study BC stemness, we developed a method to purify and amplify BC in vitro from Krox20(Cre/+), R26R(YFP/+) mouse embryos. We show that BC progeny are EGF/FGF2-responsive, form spheres, and express neural crest markers. Upon growth factor withdrawal, BC progeny gave rise to multiple neural crest and CNS lineages. Transplanted into the developing murine forebrain, they successfully survived, migrated, and integrated within the host environment. Surprisingly, BC progeny generated exclusively CNS cells, including neurons, astrocytes, and myelin-forming oligodendrocytes. In vitro experiments indicated that a sequential combination of ventralizing morphogens and glial growth factors was necessary to reprogram BC into oligodendrocytes. Thus, BC progeny are endowed with differentiation plasticity beyond the peripheral nervous system. The demonstration that CNS developmental cues can reprogram neural crest-derived stem cells into CNS derivatives suggests that BC could serve as a source of cell type-specific lineages, including oligodendrocytes, for cell-based therapies to treat CNS disorders.


Assuntos
Diferenciação Celular , Sistema Nervoso Periférico/citologia , Células-Tronco/citologia , Animais , Linhagem da Célula , Movimento Celular , Células Cultivadas , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Oligodendroglia/metabolismo
12.
Development ; 137(1): 33-42, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20023158

RESUMO

The mechanisms underlying the generation of neural cell diversity are the subject of intense investigation, which has highlighted the involvement of different signalling molecules including Shh, BMP and Wnt. By contrast, relatively little is known about FGF in this process. In this report we identify an FGF-receptor-dependent pathway in zebrafish hindbrain neural progenitors that give rise to somatic motoneurons, oligodendrocyte progenitors and differentiating astroglia. Using a combination of chemical and genetic approaches to conditionally inactivate FGF-receptor signalling, we investigate the role of this pathway. We show that FGF-receptor signalling is not essential for the survival or maintenance of hindbrain neural progenitors but controls their fate by coordinately regulating key transcription factors. First, by cooperating with Shh, FGF-receptor signalling controls the expression of olig2, a patterning gene essential for the specification of somatic motoneurons and oligodendrocytes. Second, FGF-receptor signalling controls the development of both oligodendrocyte progenitors and astroglia through the regulation of sox9, a gliogenic transcription factor the function of which we show to be conserved in the zebrafish hindbrain. Overall, for the first time in vivo, our results reveal a mechanism of FGF in the control of neural cell diversity.


Assuntos
Neurônios/citologia , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Rombencéfalo/citologia , Rombencéfalo/metabolismo , Fatores de Transcrição SOX9/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Neurônios/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
13.
Ann Neurol ; 71(5): 719-23, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22522483

RESUMO

The transcription factor EGR2 is expressed in Schwann cells, where it controls peripheral nerve myelination. Mutations of EGR2 have been found in patients with congenital hypomyelinating neuropathy or Charcot-Marie-Tooth disease type 1D. In a patient with congenital amyelinating neuropathy, we observed pathological abnormalities recapitulating the peripheral nervous system phenotype of homozygous Egr2-null mice. This patient, born from consanguineous parents, showed no EGR2 immunoreactivity in Schwann cells and harbored a homozygous 10.7-kilobase-long deletion encompassing a myelin-specific enhancer of EGR2. This regulatory mutation is the first genetic abnormality associated with congenital amyelinating neuropathy in humans.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/patologia , Proteína 2 de Resposta de Crescimento Precoce/genética , Elementos Facilitadores Genéticos/genética , Bainha de Mielina/patologia , Sequência de Bases , Feminino , Homozigoto , Humanos , Lactente , Recém-Nascido , Dados de Sequência Molecular , Linhagem , Reação em Cadeia da Polimerase , Deleção de Sequência
14.
EMBO Rep ; 12(10): 1018-23, 2011 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-21836637

RESUMO

Covalent attachment of small ubiquitin-like modifier (SUMO) to proteins regulates many processes in the eukaryotic cell. This reaction is similar to ubiquitination and usually requires an E3 ligase for substrate modification. However, only a few SUMO ligases have been described so far, which frequently facilitate sumoylation by bringing together the SUMO-conjugating enzyme Ubc9 and the target protein. Ubc9 is an interaction partner of the transcription factor Krox20, a key regulator of hindbrain development. Here, we show that Krox20 functions as a SUMO ligase for its coregulators--the Nab proteins--and that Nab sumoylation negatively modulates Krox20 transcriptional activity in vivo.


Assuntos
Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Proteínas Repressoras/metabolismo , Sumoilação , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular , Cromatina/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/genética , Regulação da Expressão Gênica , Células HEK293 , Humanos , Ligação Proteica , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina
15.
Elife ; 122023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-38095361

RESUMO

In addition to their roles in protecting nerves and increasing conduction velocity, peripheral glia plays key functions in blood vessel development by secreting molecules governing arteries alignment and maturation with nerves. Here, we show in mice that a specific, nerve-attached cell population, derived from boundary caps (BCs), constitutes a major source of mural cells for the developing skin vasculature. Using Cre-based reporter cell tracing and single-cell transcriptomics, we show that BC derivatives migrate into the skin along the nerves, detach from them, and differentiate into pericytes and vascular smooth muscle cells. Genetic ablation of this population affects the organization of the skin vascular network. Our results reveal the heterogeneity and extended potential of the BC population in mice, which gives rise to mural cells, in addition to previously described neurons, Schwann cells, and melanocytes. Finally, our results suggest that mural specification of BC derivatives takes place before their migration along nerves to the mouse skin.


Assuntos
Crista Neural , Tubo Neural , Camundongos , Animais , Crista Neural/fisiologia , Neuroglia , Células de Schwann , Pele , Diferenciação Celular/fisiologia
16.
Glia ; 60(3): 393-403, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22379615

RESUMO

The analysis of the molecular mechanisms involved in the initial interaction between neurons and Schwann cells is a key issue in understanding the myelination process. We recently identified Cthrc1 (Collagen triple helix repeat containing 1) as a gene upregulated in Schwann cells upon interaction with the axon. Cthrc1 encodes a secreted protein previously shown to be involved in migration and proliferation in different cell types. We performed a functional analysis of Cthrc1 in Schwann cells by loss-of- and gain-of-function approaches using RNA interference knockdown in cell culture and a transgenic mouse line that overexpresses the gene. This work establishes that Cthrc1 enhances Schwann cell proliferation but prevents myelination. In particular, time-course analysis of myelin formation intransgenic animals reveals that overexpression of Cthrc1 in Schwann cells leads to a delay in myelin formation with cells maintaining a proliferative state. Our data, therefore, demonstrate that Cthrc1 plays a negative regulatory role, fine-tuning the onset of peripheral myelination.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica/genética , Células de Schwann/metabolismo , Animais , Bromodesoxiuridina/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Proteína 2 de Resposta de Crescimento Precoce/genética , Embrião de Mamíferos , Proteínas da Matriz Extracelular/genética , Gânglios Espinais/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Proteína Básica da Mielina/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Células de Schwann/ultraestrutura , Fatores de Tempo , Transfecção
17.
Glia ; 60(5): 806-19, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22337526

RESUMO

The transcription factor Sox10 is expressed throughout Schwann cell development and has already been shown to be essential for specification and for the identity and further development of immature Schwann cells. Here, we show that Sox10 is also required in Schwann cells for establishing the myelinating state. This is concluded from the fact that a peripheral neuropathy develops in mice in which Sox10 is deleted by a Cre recombinase whose expression is under control of Krox20 regulatory elements. This neuropathy is characterized by altered marker gene expression along the peripheral nerve, decreased conductivity, and severe persistent hypomyelination. As the Cre recombinase is additionally active in boundary cap cells, we also analyzed the role of Sox10 during embryogenesis in establishment and maintenance of the boundary between central and peripheral nervous systems. Sox10 deletion did not affect establishment or survival of boundary cap cells but appeared to compromise barrier function as cells expressing oligodendrocyte and astrocyte markers were no longer restricted to the central nervous system, and instead found in peripheral nerves. We infer that in addition to its many roles in Schwann cells, Sox10 is also important for the integrity of the boundary between central and peripheral nervous systems.


Assuntos
Barreira Hematoencefálica/patologia , Bainha de Mielina/patologia , Doenças do Sistema Nervoso Periférico/metabolismo , Fatores de Transcrição SOXE/deficiência , Fatores de Transcrição SOXE/fisiologia , Células de Schwann/patologia , Animais , Barreira Hematoencefálica/fisiologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Bainha de Mielina/fisiologia , Sistema Nervoso Periférico/metabolismo , Sistema Nervoso Periférico/patologia , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/patologia , Fatores de Transcrição SOXE/genética , Células de Schwann/fisiologia , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia
18.
J Neurosci ; 30(17): 5958-67, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20427655

RESUMO

CNS/PNS interfaces constitute cell boundaries, because they delimit territories with different neuronal and glial contents. Despite their potential interest in regenerative medicine, the mechanisms restricting oligodendrocytes and astrocytes to the CNS and Schwann cells to the PNS in mammals are not known. To investigate the involvement of peripheral glia and myelin in the maintenance of the CNS/PNS boundary, we have first made use of different mouse mutants. We show that depletion of Schwann cells and boundary cap cells or inactivation of Krox20/Egr2, a master regulatory gene for myelination in Schwann cells, results in transgression of the CNS/PNS boundary by astrocytes and oligodendrocytes and in myelination of nerve root axons by oligodendrocytes. In contrast, such migration does not occur with the Trembler(J) mutation, which prevents PNS myelination without affecting Krox20 expression. Altogether, these data suggest that maintenance of the CNS/PNS boundary requires a Krox20 function separable from myelination control. Finally, we have analyzed a human patient affected by a congenital amyelinating neuropathy, associated with the absence of the KROX20 protein in Schwann cells. In this case, the nerve roots were also invaded by oligodendrocytes and astrocytes. This indicates that transgression of the CNS/PNS boundary by central glia can occur in pathological situations in humans and suggests that the underlying mechanisms are common with the mouse.


Assuntos
Sistema Nervoso Central/fisiologia , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Neuroglia/fisiologia , Sistema Nervoso Periférico/fisiologia , Animais , Astrócitos/fisiologia , Astrócitos/ultraestrutura , Axônios/fisiologia , Axônios/ultraestrutura , Movimento Celular/fisiologia , Sistema Nervoso Central/ultraestrutura , Proteína 2 de Resposta de Crescimento Precoce/genética , Humanos , Lactente , Camundongos , Camundongos Transgênicos , Bainha de Mielina/fisiologia , Bainha de Mielina/ultraestrutura , Neuroglia/ultraestrutura , Oligodendroglia/fisiologia , Oligodendroglia/ultraestrutura , Sistema Nervoso Periférico/ultraestrutura , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Células de Schwann/fisiologia , Células de Schwann/ultraestrutura , Raízes Nervosas Espinhais/fisiologia , Raízes Nervosas Espinhais/ultraestrutura
19.
Glia ; 59(3): 351-62, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21264944

RESUMO

The initial interaction between the Schwann cell and the axon is a complex and poorly understood aspect of the myelination process. To investigate the molecular mechanisms involved in this interaction and to identify novel genes required for myelination, we performed an RNA profiling analysis, comparing Schwann cells cultured alone or in the presence of neurons. This led to the selection of 30 genes, mostly upregulated on Schwann cell-axon interaction. Most of the identified proteins are associated with the extracellular space or signal transduction systems, consistent with possible roles in Schwann cell-axon interaction. We performed a functional analysis of one of these genes, Dok4 (downstream of kinase-4), which encodes a membrane-associated tyrosine kinase substrate. Silencing RNA-mediated knock-down of Dok4 severely affected in vitro myelination. Moreover, Dok4 is required at early stages in the myelination process, including the initial interaction with the axon, and is also involved in Schwann cell migration and proliferation. Finally, this analysis establishes the interest of our gene collection in further understanding of the molecular mechanisms involved in Schwann cell-axon interaction.


Assuntos
Axônios/metabolismo , Comunicação Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Bainha de Mielina/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Fosfoproteínas/fisiologia , Células de Schwann/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Axônios/fisiologia , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Células de Schwann/citologia
20.
J Cell Sci ; 122(Pt 18): 3340-50, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19706684

RESUMO

Stimulation of astrocytes with epidermal growth factor (EGF) induced proliferation and triggered the biosynthesis of the transcription factor Egr-1, involving the activation of the extracellular signal-regulated protein kinase (ERK) signaling pathway. No differences in the proliferation rate of astrocytes prepared from wild-type or Egr-1-deficient mice were detected. However, expression of a dominant-negative mutant of Egr-1 that interfered with DNA-binding of all Egr proteins prevented EGF-induced proliferation of astrocytes. Site-directed mutagenesis of two crucial cysteine residues within the zinc finger DNA-binding domain revealed that DNA-binding of the Egr-1 mutant was essential to inhibit proliferation of EGF-stimulated astrocytes. Expression of NAB2 (a negative co-regulator of Egr-1, Egr-2 and Egr-3) or a dominant-negative mutant of Elk-1 (a key regulator of Egr-1 biosynthesis) abolished EGF-induced proliferation of astrocytes. Chromatin immunoprecipitation experiments showed that Egr-1, Egr-2 and Egr-3 bound to the gene expressing basic fibroblast growth factor (bFGF) in EGF-stimulated astrocytes. Egr-2 and Egr-3 also interacted with the bFGF gene in EGF-stimulated astrocytes prepared from Egr-1-deficient mice, indicating that loss of Egr-1 is compensated by other Egr proteins. Together, these data show that Egr transcription factors are essential for conversion of the mitogenic signal of EGF into a proliferative response.


Assuntos
Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Fatores de Transcrição de Resposta de Crescimento Precoce/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Fator 2 Ativador da Transcrição/metabolismo , Animais , Astrócitos/enzimologia , Proliferação de Células/efeitos dos fármacos , Cromatina/metabolismo , DNA/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/deficiência , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Fatores de Transcrição de Resposta de Crescimento Precoce/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Genes Dominantes , Genes Reporter , Luciferases/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Mitógenos/farmacologia , Proteínas Mutantes/metabolismo , Mutação/genética , Proteínas de Neoplasias/metabolismo , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Elk-1 do Domínio ets/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA