Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Glia ; 68(10): 2015-2027, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32176386

RESUMO

Oligodendrocytes wrap and physically shield axons of the central nervous system with myelin sheaths, resulting in rapid signal transduction and accurate neuronal function. The complex oligodendroglial development from immature oligodendrocyte precursor cells (OPCs) to myelinating oligodendrocytes (OLs) is profoundly dependent on the activity of transcription factors of the Sox protein family. Target genes of the crucial regulator Sox10 have recently been expanded to microRNAs. Here, we report miR-204 as a novel transcriptional target of Sox10. Regulatory regions of miR-204 show responsiveness to and binding of Sox10 in reporter gene assays and electromobility shift assays. Once expressed, miR-204 inhibits OPC proliferation and facilitates differentiation into OLs in the presence of Sox10 as evident from overexpression in primary rat and mouse oligodendroglial cultures. Phenotypes are at least in part caused by miR-204-dependent repression of the pro-proliferative Ccnd2 and the differentiation inhibiting Sox4. These findings argue that the transcriptional activator Sox10 forces oligodendroglial cells to exit the cell cycle and start differentiation by gene inhibition via miR-204 induction.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , MicroRNAs/metabolismo , Oligodendroglia/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C3H , MicroRNAs/genética , Ratos
2.
Glia ; 67(5): 825-843, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30730593

RESUMO

Oligodendrocytes (OLs) facilitate information processing in the vertebrate central nervous system via axonal ensheathment. The structure and dynamics of the regulatory network that mediates oligodendrogenesis are poorly understood. We employed bioinformatics and meta-analysis of high-throughput datasets to reconstruct a regulatory network underpinning OL differentiation. From this network, we identified families of feedforward loops comprising the transcription factors (TFs) Olig2, Sox10, and Tcf7l2 and their targets. Among the targets, we found eight other TFs related to OL differentiation, suggesting a hierarchical architecture in which some TFs (Olig2, Sox10, and Tcf7l2) regulate via feedforward loops the expression of others (Sox2, Sox6, Sox11, Nkx2-2, Nkx6-2, Hes5, Myt1, and Myrf). Model simulations with a kinetic model reproduced the mechanisms of OL differentiation only when in the model, Sox10-mediated repression of Tcf7l2 by miR-338/miR-155 was introduced, a prediction confirmed in genetic functional experiments. Additional model simulations suggested that OLs from dorsal regions emerge through BMP/Sox9 signaling.


Assuntos
Diferenciação Celular/fisiologia , Redes Reguladoras de Genes , Modelos Biológicos , Dinâmica não Linear , Oligodendroglia/fisiologia , Animais , Simulação por Computador , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Nucleares , Fatores de Transcrição
3.
Int J Mol Sci ; 20(11)2019 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-31159496

RESUMO

In the central nervous system, oligodendrocytes wrap axons with myelin sheaths, which is essential for rapid transfer of electric signals and their trophic support. In oligodendroglia, transcription factors of the Sox protein family are pivotal regulators of a variety of developmental processes. These include specification, proliferation, and migration of oligodendrocyte precursor cells as well as terminal differentiation to mature myelinating oligodendrocytes. Sox proteins are further affected in demyelinating diseases and are involved in remyelination following damage of the central nervous system. Here we summarize and discuss latest findings on transcriptional regulation of Sox proteins, their function, target genes, and interaction with other transcription factors and chromatin remodelers in oligodendroglia with physiological and pathophysiological relevance.


Assuntos
Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Fatores de Transcrição SOX/metabolismo , Animais , Montagem e Desmontagem da Cromatina , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/metabolismo , Regulação da Expressão Gênica , Humanos , Bainha de Mielina/genética , Oligodendroglia/citologia , Fatores de Transcrição SOX/genética
4.
Glia ; 66(2): 279-294, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29023979

RESUMO

In Schwann cells of the vertebrate peripheral nervous system, induction of myelination and myelin maintenance both depend on the HMG-domain-containing transcription factor Sox10. In oligodendrocytes of the central nervous system, Sox10 is also essential for the induction of myelination. Its role in late phases of myelination and myelin maintenance has not been studied so far. Here, we show that these processes are largely unaffected in mice that lack Sox10 in mature oligodendrocytes. As Sox10 is co-expressed with the related Sox8, we also analyzed oligodendrocytes and myelination in Sox8-deficient mice. Again, we could not detect any major abnormalities. Expression of many myelin genes was only modestly reduced in both mouse mutants. Dramatic reductions in expression levels and phenotypic disturbances became only apparent once Sox8 and Sox10 were both absent. This argues that Sox8 and Sox10 are jointly required for myelin maintenance and impact myelin gene expression. One direct target gene of both Sox proteins is the late myelin gene Mog. Our results point to at least partial functional redundancy between both related Sox proteins in mature oligodendrocytes and are the first report of a substantial function of Sox8 in the oligodendroglial lineage.


Assuntos
Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Fatores de Transcrição SOXE/biossíntese , Células de Schwann/metabolismo , Animais , Linhagem da Célula , Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Fatores de Transcrição SOXE/genética
5.
J Neurochem ; 146(3): 251-268, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29749639

RESUMO

The high-mobility-group domain containing SoxC transcription factors Sox4 and Sox11 are expressed and required in the vertebrate central nervous system in neuronal precursors and neuroblasts. To identify genes that are widely regulated by SoxC proteins during vertebrate neurogenesis we generated expression profiles from developing mouse brain and chicken neural tube with reduced SoxC expression and found the transcription factor prospero homeobox protein 1 (Prox1) strongly down-regulated under both conditions. This led us to hypothesize that Prox1 expression depends on SoxC proteins in the developing central nervous system of mouse and chicken. By combining luciferase reporter assays and over-expression in the chicken neural tube with in vivo and in vitro binding studies, we identify the Prox1 gene promoter and two upstream enhancers at -44 kb and -40 kb relative to the transcription start as regulatory regions that are bound and activated by SoxC proteins. This argues that Prox1 is a direct target gene of SoxC proteins during neurogenesis. Electroporations in the chicken neural tube furthermore show that Prox1 activates a subset of SoxC target genes, whereas it has no effects on others. We propose that the transcriptional control of Prox1 by SoxC proteins may ensure coupling of two types of transcription factors that are both required during early neurogenesis, but have at least in part distinct functions. Open Data: Materials are available on https://cos.io/our-services/open-science-badges/ https://osf.io/93n6m/.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Prosencéfalo/citologia , Fatores de Transcrição SOXC/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Embrião de Galinha , Imunoprecipitação da Cromatina , Biologia Computacional , Ensaio de Desvio de Mobilidade Eletroforética , Eletroporação , Embrião de Mamíferos , Ontologia Genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Tubo Neural/citologia , Tubo Neural/metabolismo , Fatores do Domínio POU/genética , Fatores do Domínio POU/metabolismo , Prosencéfalo/embriologia , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Fatores de Transcrição SOXC/genética , Tubulina (Proteína)/metabolismo , Proteínas Supressoras de Tumor/genética
6.
Glia ; 65(7): 1089-1102, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28370559

RESUMO

During development of myelin-forming oligodendrocytes in the central nervous system the two closely related transcription factors Sox9 and Sox10 play essential roles that are partly shared and partly unique. Whereas Sox9 primarily functions during oligodendroglial specification, Sox10 is uniquely required to induce terminal differentiation and myelination. During this process, Sox10 protein levels rise substantially. As this coincides with a reciprocal decrease in Sox9, we postulated that Sox10 influences Sox9 amounts in differentiating oligodendrocytes. Here we show that Sox9 levels are indeed inversely coupled to Sox10 levels such that Sox10 deletion in oligodendroglial cells evokes a reciprocal increase in Sox9. We furthermore provide evidence that this coupling involves upregulation of microRNAs miR335 and miR338 as direct transcriptional targets of Sox10. The two microRNAs in turn recognize the 3'-UTR of Sox9 mRNA and may thereby reduce Sox9 protein levels posttranscriptionally in oligodendroglial cells. Such a mechanism may enable oligodendroglial cells to adapt the ratio of both related Sox proteins in a manner required for successful lineage progression and differentiation. Mathematical modeling furthermore shows that the identified regulatory circuit has the potential to convert a transient stimulus into an irreversible switch of cellular properties and may thus contribute to terminal differentiation of oligodendrocytes.


Assuntos
Regulação da Expressão Gênica/genética , MicroRNAs/metabolismo , Oligodendroglia/metabolismo , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOXE/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Células Cultivadas , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Modelos Biológicos , Modelos Moleculares , Modelos Teóricos , Proteína Básica da Mielina/metabolismo , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Ratos , Fatores de Transcrição SOXE/genética , Fatores de Transcrição/metabolismo , Transfecção
7.
Development ; 141(1): 39-50, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24257626

RESUMO

Neural precursor cells of the ventricular zone give rise to all neurons and glia of the central nervous system and rely for maintenance of their precursor characteristics on the closely related SoxB1 transcription factors Sox1, Sox2 and Sox3. We show in mouse spinal cord that, whereas SoxB1 proteins are usually downregulated upon neuronal specification, they continue to be expressed in glial precursors. In the oligodendrocyte lineage, Sox2 and Sox3 remain present into the early phases of terminal differentiation. Surprisingly, their deletion does not alter precursor characteristics but interferes with proper differentiation. Although a direct influence on myelin gene expression may be part of their function, we provide evidence for another mode of action. SoxB1 proteins promote oligodendrocyte differentiation in part by negatively controlling miR145 and thereby preventing this microRNA from inhibiting several pro-differentiation factors. This study presents one of the few cases in which SoxB1 proteins, including the stem cell factor Sox2, are associated with differentiation rather than precursor functions.


Assuntos
MicroRNAs/genética , Oligodendroglia/metabolismo , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Camundongos , Células-Tronco Neurais , Neurogênese , Neuroglia/citologia , Neuroglia/metabolismo , Regiões Promotoras Genéticas , Ratos , Fatores de Transcrição SOX9/biossíntese , Medula Espinal/citologia , Medula Espinal/embriologia , Medula Espinal/metabolismo
8.
Acta Neuropathol ; 132(1): 59-75, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27059609

RESUMO

Multiple system atrophy (MSA) is a rare atypical parkinsonian disorder characterized by a rapidly progressing clinical course and at present without any efficient therapy. Neuropathologically, myelin loss and neurodegeneration are associated with α-synuclein accumulation in oligodendrocytes, but underlying pathomechanisms are poorly understood. Here, we analyzed the impact of oligodendrocytic α-synuclein on the formation of myelin sheaths to define a potential interventional target for MSA. Post-mortem analyses of MSA patients and controls were performed to quantify myelin and oligodendrocyte numbers. As pre-clinical models, we used transgenic MSA mice, a myelinating stem cell-derived oligodendrocyte-neuron co-culture, and primary oligodendrocytes to determine functional consequences of oligodendrocytic α-synuclein overexpression on myelination. We detected myelin loss accompanied by preserved or even increased numbers of oligodendrocytes in post-mortem MSA brains or transgenic mouse forebrains, respectively, indicating an oligodendrocytic dysfunction in myelin formation. Corroborating this observation, overexpression of α-synuclein in primary and stem cell-derived oligodendrocytes severely impaired myelin formation, defining a novel α-synuclein-linked pathomechanism in MSA. We used the pro-myelinating activity of the muscarinic acetylcholine receptor antagonist benztropine to analyze the reversibility of the myelination deficit. Transcriptome profiling of primary pre-myelinating oligodendrocytes demonstrated that benztropine readjusts myelination-related processes such as cholesterol and membrane biogenesis, being compromised by oligodendrocytic α-synuclein. Additionally, benztropine restored the α-synuclein-induced myelination deficit of stem cell-derived oligodendrocytes. Strikingly, benztropine also ameliorated the myelin deficit in transgenic MSA mice, resulting in a prevention of neuronal cell loss. In conclusion, this study defines the α-synuclein-induced myelination deficit as a novel and crucial pathomechanism in MSA. Importantly, the reversible nature of this oligodendrocytic dysfunction opens a novel avenue for an intervention in MSA.


Assuntos
Antiparkinsonianos/farmacologia , Benzotropina/farmacologia , Atrofia de Múltiplos Sistemas/tratamento farmacológico , Atrofia de Múltiplos Sistemas/metabolismo , alfa-Sinucleína/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Gliose/metabolismo , Gliose/patologia , Gliose/prevenção & controle , Masculino , Camundongos Transgênicos , Atrofia de Múltiplos Sistemas/diagnóstico por imagem , Atrofia de Múltiplos Sistemas/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Ratos Wistar , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/patologia , Transcriptoma/efeitos dos fármacos , alfa-Sinucleína/genética
9.
Cell Tissue Res ; 359(1): 111-24, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24894327

RESUMO

Neuroepithelial precursor cells of the vertebrate central nervous system either self-renew or differentiate into neurons, oligodendrocytes or astrocytes under the influence of a gene regulatory network that consists in transcription factors, epigenetic modifiers and microRNAs. Sox transcription factors are central to this regulatory network, especially members of the SoxB, SoxC, SoxD, SoxE and SoxF groups. These Sox proteins are widely expressed in neuroepithelial precursor cells and in newly specified, differentiating and mature neurons, oligodendrocytes and astrocytes and influence their identity, survival and development. They exert their effect predominantly at the transcriptional level but also have substantial impact on expression at the epigenetic and posttranscriptional levels with some Sox proteins acting as pioneer factors, recruiting chromatin-modifying and -remodelling complexes or influencing microRNA expression. They interact with a large variety of other transcription factors and influence the expression of regulatory molecules and effector genes in a cell-type-specific and temporally controlled manner. As versatile regulators with context-dependent functions, they are not only indispensable for central nervous system development but might also be instrumental for the development of reprogramming and cell conversion strategies for replacement therapies and for assisted regeneration after injury or degeneration-induced cell loss in the central nervous system.


Assuntos
Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Fatores de Transcrição SOX/metabolismo , Células-Tronco/metabolismo , Animais , Humanos , Neurogênese , Neuroglia/citologia , Neurônios/citologia , Fatores de Transcrição SOX/química , Células-Tronco/citologia
10.
Mol Cell Neurosci ; 62: 68-78, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25019582

RESUMO

Myelin loss is a widespread neuropathological hallmark of the atypical parkinsonian disorder multiple system atrophy (MSA). On a cellular level, MSA is characterized by alpha-synuclein (aSyn)-positive glial cytoplasmic inclusions (GCIs) within mature oligodendrocytes leading to demyelination as well as axonal and neuronal loss. Oligodendrocyte progenitor cells (OPCs) represent a proliferative cell population distributed throughout the adult mammalian central nervous system. During remyelination, OPCs are recruited to sites of demyelination, differentiate, and finally replace dysfunctional mature oligodendrocytes. However, comprehensive studies investigating OPCs and remyelination processes in MSA are lacking. In the present study, we therefore investigate the effect of human aSyn (h-aSyn) on early primary rat OPC maturation. Upon lentiviral transduction, h-aSyn expressing OPCs exhibit fewer and shorter primary processes at the initiation of differentiation. Until day 4 of a 6day differentiation paradigm, h-aSyn expressing OPCs further show a severely delayed maturation evidenced by reduced myelin gene expression and increased levels of the progenitor marker platelet derived growth factor receptor-alpha (PDGFRα). Matching these results, OPCs that take up extracellular recombinant h-aSyn exhibit a similar delayed differentiation. In both experimental setups however, myelin gene expression is restored at day 6 of differentiation paralleled by decreased intracellular h-aSyn levels indicating a reverse correlation of h-aSyn and the differentiation potential of OPCs. Taken together, these findings suggest a tight link between the intracellular level of h-aSyn and maturation capacity of primary OPCs.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , alfa-Sinucleína/metabolismo , Animais , Axônios/metabolismo , Células Cultivadas , Doenças Desmielinizantes/metabolismo , Espaço Intracelular/metabolismo , Proteína Básica da Mielina/metabolismo , Bainha de Mielina/metabolismo , Ratos Wistar
11.
J Neurosci ; 33(15): 6679-90, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23575864

RESUMO

Several transcription factors are essential for terminal differentiation of myelinating glia, among them the high-mobility-group-domain-containing protein Sox10. To better understand how these factors exert their effects and shape glial expression programs, we identified and characterized a physical and functional link between Sox10 and the Med12 subunit of the Mediator complex that serves as a conserved multiprotein interphase between transcription factors and the general transcription machinery. We found that Sox10 bound with two of its conserved domains to the C-terminal region of Med12 and its close relative, Med12-like. In contrast to Med12-like, substantial amounts of Med12 were detected in both Schwann cells and oligodendrocytes. Its conditional glia-specific deletion in mice led to terminal differentiation defects that were highly reminiscent of those obtained after Sox10 deletion. In support of a functional cooperation, both proteins were jointly required for Krox20 induction and were physically associated with the critical regulatory region of the Krox20 gene in myelinating Schwann cells. We conclude that Sox10 functions during terminal differentiation of myelinating glia, at least in part by Med12-dependent recruitment of the Mediator complex.


Assuntos
Diferenciação Celular/fisiologia , Complexo Mediador/fisiologia , Oligodendroglia/citologia , Fatores de Transcrição SOXE/fisiologia , Células de Schwann/citologia , Animais , Diferenciação Celular/genética , Linhagem Celular , Proteína 2 de Resposta de Crescimento Precoce/biossíntese , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Masculino , Complexo Mediador/genética , Camundongos , Camundongos Transgênicos , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Ligação Proteica/genética , Ligação Proteica/fisiologia , Fatores de Transcrição SOXE/genética , Células de Schwann/metabolismo
12.
Nucleic Acids Res ; 40(1): 88-101, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21908409

RESUMO

The Sox10 transcription factor is a central regulator of vertebrate neural crest and nervous system development. Its expression is likely controlled by multiple enhancer elements, among them U3 (alternatively known as MCS4). Here we analyze U3 activity to obtain deeper insights into Sox10 function and expression in the neural crest and its derivatives. U3 activity strongly depends on the presence of Sox10 that regulates its own expression as commonly observed for important developmental regulators. Sox10 bound directly as monomer to at least three sites in U3, whereas a fourth site preferred dimers. Deletion of these sites efficiently reduced U3 activity in transfected cells and transgenic mice. In stimulating the U3 enhancer, Sox10 synergized with many other transcription factors present in neural crest and developing peripheral nervous system including Pax3, FoxD3, AP2α, Krox20 and Sox2. In case of FoxD3, synergism involved Sox10-dependent recruitment to the U3 enhancer, while Sox10 and AP2α each had to bind to the regulatory region. Our study points to the importance of autoregulatory activity and synergistic interactions for maintenance of Sox10 expression and functional activity of Sox10 in the neural crest regulatory network.


Assuntos
Elementos Facilitadores Genéticos , Crista Neural/metabolismo , Fatores de Transcrição SOXE/metabolismo , Ativação Transcricional , Animais , Sítios de Ligação , Embrião de Galinha , Células HEK293 , Homeostase , Humanos , Camundongos , Camundongos Transgênicos , Neuroglia/metabolismo , Ratos , Fatores de Transcrição SOX/metabolismo , Fatores de Transcrição SOXE/genética , Fatores de Transcrição/metabolismo
13.
Cells ; 13(11)2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38891067

RESUMO

Rapid information processing in the central nervous system requires the myelination of axons by oligodendrocytes. The transcription factor Sox2 and its close relative Sox3 redundantly regulate the development of myelin-forming oligodendrocytes, but little is known about the underlying molecular mechanisms. Here, we characterized the expression profile of cultured oligodendroglial cells during early differentiation and identified Bcas1, Enpp6, Zfp488 and Nkx2.2 as major downregulated genes upon Sox2 and Sox3 deletion. An analysis of mice with oligodendrocyte-specific deletion of Sox2 and Sox3 validated all four genes as downstream targets in vivo. Additional functional assays identified regulatory regions in the vicinity of each gene that are responsive to and bind both Sox proteins. Bcas1, Enpp6, Zfp488 and Nkx2.2 therefore likely represent direct target genes and major effectors of Sox2 and Sox3. Considering the preferential expression and role of these genes in premyelinating oligodendrocytes, our findings suggest that Sox2 and Sox3 impact oligodendroglial development at the premyelinating stage with Bcas1, Enpp6, Zfp488 and Nkx2.2 as their major effectors.


Assuntos
Diferenciação Celular , Proteína Homeobox Nkx-2.2 , Oligodendroglia , Fatores de Transcrição SOXB1 , Fatores de Transcrição , Animais , Camundongos , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Oligodendroglia/metabolismo , Oligodendroglia/citologia , Diester Fosfórico Hidrolases/metabolismo , Diester Fosfórico Hidrolases/genética , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXC/metabolismo , Fatores de Transcrição SOXC/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética
14.
Biol Chem ; 394(12): 1583-93, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23729567

RESUMO

Sox10 belongs to the Sox family of high-mobility group-box transcription factors. It fulfils widespread and essential functions in myelinating glia at multiple stages of development such as glial specification, survival and terminal differentiation. To a large extent, these diverse activities can be attributed to its capacity to interact with different transcription factors in distinct regulatory networks. Beyond transcription factors, an increasing number of interaction partners are emerging with alternative impact on gene expression. These include components of the mediator complex, the Brahma-associated factor complex and histone deacetylases. Here, we discuss interactions with functional relevance in myelinating glia and link Sox10 function in these cells not only to gene transcription, but also to epigenetics and chromatin remodeling.


Assuntos
Epigênese Genética , Bainha de Mielina/metabolismo , Neuroglia/metabolismo , Fatores de Transcrição SOXE/metabolismo , Transcrição Gênica , Animais , Montagem e Desmontagem da Cromatina , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Humanos , Complexo Mediador/metabolismo , Oligodendroglia/metabolismo , Fatores de Transcrição SOXE/genética , Células de Schwann/metabolismo
15.
Dev Biol ; 332(2): 418-28, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19527706

RESUMO

Related transcription factors of the POU protein family show extensive overlap of expression in vivo and exhibit very similar biochemical properties in vitro. To study functional equivalence of class III POU proteins in vivo, we exchanged the Oct-6 gene by Brn-1 in the mouse. Brn-1 can fully replace Oct-6 in Schwann cells and rescue peripheral nervous system development in these mice. The same mice, however, exhibit severe defects in forebrain development arguing that Oct-6 and Brn-1 are not functionally equivalent in the central nervous system. The cause of the observed forebrain phenotype is complex, but anteriorly expanded Wnt1 expression contributes. Oct-6 normally represses Wnt1 expression in the early diencephalon and replacement by Brn-1 as a weaker inhibitor is no longer sufficient to maintain the necessary level of repression in the mouse mutant. The extent of functional equivalence between related transcription factors is thus strongly dependent on the analyzed tissue.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Fatores do Domínio POU/metabolismo , Prosencéfalo , Animais , Biomarcadores/metabolismo , Linhagem Celular , Humanos , Hibridização In Situ , Mesencéfalo/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Crista Neural/fisiologia , Fator 6 de Transcrição de Octâmero/genética , Fatores do Domínio POU/genética , Fenótipo , Prosencéfalo/anormalidades , Prosencéfalo/embriologia , Rombencéfalo/embriologia , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
16.
J Neurochem ; 112(3): 744-54, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19922439

RESUMO

The high-mobility group domain transcription factor Sox10 is believed to influence myelination in Schwann cells by directly activating myelin genes and by inducing Krox20 as a pivotal regulator of peripheral myelination. Krox20 induction at this stage is thought to be mediated by the myelinating Schwann cell element 35 kb downstream of the Krox20 transcriptional start site and requires cooperation with Oct6. Here, we prove for the first time in vivo that Schwann cell-specific Krox20 expression indeed depends on Sox10. We also provide evidence that Sox10 functions through multiple, mostly monomeric binding sites in the myelinating Schwann cell element in a manner that should render the enhancer exquisitely sensitive to Sox10 levels. Synergistic activation of the enhancer by Sox10 and Oct6 furthermore does not involve cooperative binding to closely spaced binding sites in defined composite elements. Nevertheless, the POU domain of Oct6 and the high-mobility group domain of Sox10 as the two DNA-binding domains were both essential indicating that each transcription factor has to bind independently to DNA. Whereas the POU domain was the only important region of Oct6, two further Sox10 domains were required for synergistic Krox20 activation. These were the carboxyterminal transactivation domain and the conserved K2 domain in the central portion of Sox10. All required regions are conserved in several closely related POU and Sox proteins thus explaining why Oct6 and Sox10 can be replaced by their relatives during Krox20 induction in myelinating Schwann cells.


Assuntos
Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fatores de Transcrição SOXE/metabolismo , Células de Schwann/metabolismo , Animais , Sítios de Ligação/genética , Linhagem Celular Transformada , Proteína 2 de Resposta de Crescimento Precoce/genética , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Fatores de Transcrição SOXE/química , Fatores de Transcrição SOXE/genética , Transfecção/métodos
17.
Sci Rep ; 10(1): 17807, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33082503

RESUMO

The high mobility group-domain containing transcription factor Sox10 is an essential regulator of developmental processes and homeostasis in the neural crest, several neural crest-derived lineages and myelinating glia. Recent studies have also implicated Sox10 as an important factor in mammary stem and precursor cells. Here we employ a series of mouse mutants with constitutive and conditional Sox10 deficiencies to show that Sox10 has multiple functions in the developing mammary gland. While there is no indication for a requirement of Sox10 in the specification of the mammary placode or descending mammary bud, it is essential for both the prenatal hormone-independent as well as the pubertal hormone-dependent branching of the mammary epithelium and for proper alveologenesis during pregnancy. It furthermore acts in a dosage-dependent manner. Sox10 also plays a role during the involution process at the end of the lactation period. Whereas its effect on epithelial branching and alveologenesis are likely causally related to its function in mammary stem and precursor cells, this is not the case for its function during involution where Sox10 seems to work at least in part through regulation of the miR-424(322)/503 cluster.


Assuntos
Epitélio/fisiologia , Glândulas Mamárias Animais/fisiologia , Morfogênese/fisiologia , Crista Neural/fisiologia , Fatores de Transcrição SOXE/metabolismo , Animais , Diferenciação Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Homeostase , Lactação , Camundongos , Camundongos Transgênicos , MicroRNAs/genética , Mutação/genética , Fatores de Transcrição SOXE/genética
18.
Brain Pathol ; 29(3): 380-396, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30444295

RESUMO

Neuroinflammation and oligodendroglial cytoplasmic α-synuclein (α-syn) inclusions (GCIs) are important neuropathological characteristics of multiple system atrophy (MSA). GCIs are known to interfere with oligodendroglial maturation and consequently result in myelin loss. The neuroinflammatory phenotype in the context of MSA, however, remains poorly understood. Here, we demonstrate MSA-associated neuroinflammation being restricted to myeloid cells and tightly linked to oligodendroglial α-syncleinopathy. In human putaminal post-mortem tissue of MSA patients, neuroinflammation was observed in white matter regions only. This locally restricted neuroinflammation coincided with elevated numbers of α-syn inclusions, while gray matter with less α-synucleinopathy remained unaffected. In order to analyze the temporal pattern of neuroinflammation, a transgenic mouse model overexpressing human α-syn under the control of an oligodendrocyte-specific myelin basic protein (MBP) promoter (MBP29-hα-syn mice) was assessed in a pre-symptomatic and symptomatic disease stage. Strikingly, we detected an increased neuroinflammation in regions with a high α-syn load, the corpus callosum and the striatum, of MBP29-hα-syn mice, already at a pre-symptomatic stage. Furthermore, this inflammatory response was restricted to myeloid cells being highly proliferative and showing an activated, phagocytic phenotype. In contrast, severe astrogliosis was observed only in gray matter regions of MSA patients as well as MBP29-hα-syn mice. To further characterize the influence of oligodendrocytes on initiation of the myeloid immune response, we performed RNA sequencing analysis of α-syn overexpressing primary oligodendrocytes. A distinct gene expression profile including upregulation of cytokines important for myeloid cell attraction and proliferation was detected in α-syn overexpressing oligodendrocytes. Additionally, microdissected tissue of MBP29-hα-syn mice exhibited a similar cellular gene expression profile in white matter regions even pre-symptomatically. Collectively, these results imply an early crosstalk between neuroinflammation and oligodendrocytes containing α-syn inclusions leading to an immune response locally restricted to white matter regions in MSA.


Assuntos
Atrofia de Múltiplos Sistemas/fisiopatologia , Oligodendroglia/patologia , Sinucleinopatias/metabolismo , Idoso , Animais , Encéfalo/patologia , Corpo Estriado/patologia , Modelos Animais de Doenças , Feminino , Humanos , Corpos de Inclusão/patologia , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Atrofia de Múltiplos Sistemas/metabolismo , Células Mieloides/metabolismo , Neuroimunomodulação/fisiologia , Neurônios/patologia , Oligodendroglia/metabolismo , Sinucleinopatias/imunologia , Substância Branca/patologia , alfa-Sinucleína/metabolismo
19.
Neurogenesis (Austin) ; 1(1): e962391, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-27502481

RESUMO

The transcription factor Sox2 is best known as a pluripotency factor in stem and precursor cells and its expression generally correlates with an undifferentiated state. Proposed modes of action include those as classical transcription factor and pre-patterning factor with influence on histone modifications and chromatin structure. Recently, we provided the first detailed analysis of Sox2 expression and function during development of oligodendrocytes, the myelin-forming cells of the CNS. Surprisingly, we found evidence for a role of Sox2 as differentiation factor and found it to act through modulation of microRNA levels. Thus, we add new facets to the functional repertoire of Sox2 and throw light on the networking activity of this multitasking developmental regulator.

20.
PLoS One ; 9(4): e94580, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24718611

RESUMO

Sensory nerves of the brainstem are mostly composed of placode-derived neurons, neural crest-derived neurons and neural crest-derived Schwann cells. This mixed origin of cells has made it difficult to dissect interdependence for fiber guidance. Inner ear-derived neurons are known to connect to the brain after delayed loss of Schwann cells in ErbB2 mutants. However, the ErbB2 mutant related alterations in the ear and the brain compound interpretation of the data. We present here a new model to evaluate exclusively the effect of Schwann cell loss on inner ear innervation. Conditional deletion of the neural crest specific transcription factor, Sox10, using the rhombic lip/neural crest specific Wnt1-cre driver spares Sox10 expression in the ear. We confirm that neural crest-derived cells provide a stop signal for migrating spiral ganglion neurons. In the absence of Schwann cells, spiral ganglion neurons migrate into the center of the cochlea and even out of the ear toward the brain. Spiral ganglion neuron afferent processes reach the organ of Corti, but many afferent fibers bypass the organ of Corti to enter the lateral wall of the cochlea. In contrast to this peripheral disorganization, the central projection to cochlear nuclei is normal. Compared to ErbB2 mutants, conditional Sox10 mutants have limited cell death in spiral ganglion neurons, indicating that the absence of Schwann cells alone contributes little to the embryonic survival of neurons. These data suggest that neural crest-derived cells are dispensable for all central and some peripheral targeting of inner ear neurons. However, Schwann cells provide a stop signal for migratory spiral ganglion neurons and facilitate proper targeting of the organ of Corti by spiral ganglion afferents.


Assuntos
Movimento Celular , Orelha Interna/citologia , Deleção de Genes , Marcação de Genes , Neurônios/citologia , Fatores de Transcrição SOXE/metabolismo , Proteína Wnt1/metabolismo , Animais , Apoptose , Feminino , Integrases/metabolismo , Metaloproteinases da Matriz Associadas à Membrana/metabolismo , Camundongos Knockout , Modelos Biológicos , Mutação/genética , Fatores de Crescimento Neural/metabolismo , Órgão Espiral/citologia , Recombinação Genética/genética , Reprodutibilidade dos Testes , Células de Schwann/citologia , Células de Schwann/metabolismo , Gânglio Espiral da Cóclea/citologia , Vestíbulo do Labirinto/citologia , Vestíbulo do Labirinto/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA