Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 42(10): 113272, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37858465

RESUMO

Remyelination after white matter injury (WMI) often fails in diseases such as multiple sclerosis because of improper recruitment and repopulation of oligodendrocyte precursor cells (OPCs) in lesions. How OPCs elicit specific intracellular programs in response to a chemically and mechanically diverse environment to properly regenerate myelin remains unclear. OPCs construct primary cilia, specialized signaling compartments that transduce Hedgehog (Hh) and G-protein-coupled receptor (GPCR) signals. We investigated the role of primary cilia in the OPC response to WMI. Removing cilia from OPCs genetically via deletion of Ift88 results in OPCs failing to repopulate WMI lesions because of reduced proliferation. Interestingly, loss of cilia does not affect Hh signaling in OPCs or their responsiveness to Hh signals but instead leads to dysfunctional cyclic AMP (cAMP)-dependent cAMP response element-binding protein (CREB)-mediated transcription. Because inhibition of CREB activity in OPCs reduces proliferation, we propose that a GPCR/cAMP/CREB signaling axis initiated at OPC cilia orchestrates OPC proliferation during development and in response to WMI.


Assuntos
Células Precursoras de Oligodendrócitos , Substância Branca , Células Precursoras de Oligodendrócitos/metabolismo , Cílios/metabolismo , Substância Branca/metabolismo , Proteínas Hedgehog/metabolismo , Oligodendroglia/metabolismo , Bainha de Mielina/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proliferação de Células , Diferenciação Celular/fisiologia
2.
Neuron ; 111(2): 190-201.e8, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36384142

RESUMO

Oligodendrocyte precursor cells (OPCs) undergo an extensive and coordinated migration in the developing CNS, using the pre-formed scaffold of developed blood vessels as their physical substrate for migration. While OPC association with vasculature is critical for dispersal, equally important for permitting differentiation and proper myelination of target axons is their appropriate and timely detachment, but regulation of this process remains unclear. Here we demonstrate a correlation between the developmental formation of astrocytic endfeet on vessels and the termination of OPC perivascular migration. Ex vivo and in vivo live imaging shows that astrocyte endfeet physically displace OPCs from vasculature, and genetic abrogation of endfoot formation hinders both OPC detachment from vessels and subsequent differentiation. Astrocyte-derived semaphorins 3a and 6a act to repel OPCs from blood vessels at the cessation of their perivascular migration and, in so doing, permit subsequent OPC differentiation by insulating them from a maturation inhibitory endothelial niche.


Assuntos
Células Precursoras de Oligodendrócitos , Astrócitos , Oligodendroglia/fisiologia , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia
3.
Nat Neurosci ; 25(11): 1528-1542, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36303069

RESUMO

Astrocytes become reactive in response to insults to the central nervous system by adopting context-specific cellular signatures and outputs, but a systematic understanding of the underlying molecular mechanisms is lacking. In this study, we developed CRISPR interference screening in human induced pluripotent stem cell-derived astrocytes coupled to single-cell transcriptomics to systematically interrogate cytokine-induced inflammatory astrocyte reactivity. We found that autocrine-paracrine IL-6 and interferon signaling downstream of canonical NF-κB activation drove two distinct inflammatory reactive signatures, one promoted by STAT3 and the other inhibited by STAT3. These signatures overlapped with those observed in other experimental contexts, including mouse models, and their markers were upregulated in human brains in Alzheimer's disease and hypoxic-ischemic encephalopathy. Furthermore, we validated that markers of these signatures were regulated by STAT3 in vivo using a mouse model of neuroinflammation. These results and the platform that we established have the potential to guide the development of therapeutics to selectively modulate different aspects of inflammatory astrocyte reactivity.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Humanos , Astrócitos , Transdução de Sinais , Citocinas , Inflamação
4.
Dev Neurobiol ; 81(8): 985-996, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34643996

RESUMO

Oligodendrocytes, the myelinating cells of the central nervous system (CNS), develop from oligodendrocyte progenitor cells (OPCs) that must first migrate extensively throughout the developing brain and spinal cord. Specified at particular times from discrete regions in the developing CNS, OPCs are one of the most migratory of cell types and disperse rapidly. A variety of factors act on OPCs to trigger intracellular changes that regulate their migration. We will discuss factors that act as long-range guidance cues, those that act to regulate cellular motility, and those that are critical in determining the final positioning of OPCs. In addition, recent evidence has identified the vasculature as the physical substrate used by OPCs for their migration. Several new findings relating to this oligodendroglial-vascular signaling axis reveal new insight on the relationship between OPCs and blood vessels in the developing and adult brain.


Assuntos
Células Precursoras de Oligodendrócitos , Diferenciação Celular/fisiologia , Sistema Nervoso Central , Células Precursoras de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Medula Espinal
5.
Neuron ; 109(19): 3104-3118.e6, 2021 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-34390652

RESUMO

Oligodendrocyte (OL) maturation arrest in human white matter injury contributes significantly to the failure of endogenous remyelination in multiple sclerosis (MS) and newborn brain injuries such as hypoxic ischemic encephalopathy (HIE) that cause cerebral palsy. In this study, we identify an oligodendroglial-intrinsic factor that controls OL maturation specifically in the setting of injury. We find a requirement for the ring finger protein Rnf43 not in normal development but in neonatal hypoxic injury and remyelination in the adult mammalian CNS. Rnf43, but not the related Znrf3, is potently activated by Wnt signaling in OL progenitor cells (OPCs) and marks activated OPCs in human MS and HIE. Rnf43 is required in an injury-specific context, and it promotes OPC differentiation through negative regulation of Wnt signal strength in OPCs at the level of Fzd1 receptor presentation on the cell surface. Inhibition of Fzd1 using UM206 promotes remyelination following ex vivo and in vivo demyelinating injury.


Assuntos
Lesões Encefálicas/genética , Lesões Encefálicas/patologia , Oligodendroglia/patologia , Ubiquitina-Proteína Ligases/genética , Animais , Lesões Encefálicas/metabolismo , Doenças Desmielinizantes/genética , Receptores Frizzled/efeitos dos fármacos , Receptores Frizzled/genética , Humanos , Camundongos , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/fisiologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Remielinização/efeitos dos fármacos , Remielinização/genética , Células-Tronco/metabolismo , Células-Tronco/patologia , Substância Branca/metabolismo , Substância Branca/patologia , Via de Sinalização Wnt
6.
Nat Neurosci ; 24(2): 234-244, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33526922

RESUMO

Fibrosis is a common pathological response to inflammation in many peripheral tissues and can prevent tissue regeneration and repair. Here, we identified persistent fibrotic scarring in the CNS following immune cell infiltration in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis. Using lineage tracing and single-cell sequencing in EAE, we determined that the majority of the fibrotic scar is derived from proliferative CNS fibroblasts, not pericytes or infiltrating bone marrow-derived cells. Ablating proliferating fibrotic cells using cell-specific expression of herpes thymidine kinase led to an increase in oligodendrocyte lineage cells within the inflammatory lesions and a reduction in motor disability. We further identified that interferon-gamma pathway genes are enriched in CNS fibrotic cells, and the fibrotic cell-specific deletion of Ifngr1 resulted in reduced fibrotic scarring in EAE. These data delineate a framework for understanding the CNS fibrotic response.


Assuntos
Barreira Hematoencefálica/patologia , Encefalomielite Autoimune Experimental/patologia , Fibroblastos/patologia , Fibrose/patologia , Infiltração de Neutrófilos , Medula Espinal/patologia , Animais , Camundongos , Oligodendroglia/patologia
7.
Nat Neurosci ; 22(5): 709-718, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30988524

RESUMO

Disruption of the blood-brain barrier (BBB) is critical to initiation and perpetuation of disease in multiple sclerosis (MS). We report an interaction between oligodendroglia and vasculature in MS that distinguishes human white matter injury from normal rodent demyelinating injury. We find perivascular clustering of oligodendrocyte precursor cells (OPCs) in certain active MS lesions, representing an inability to properly detach from vessels following perivascular migration. Perivascular OPCs can themselves disrupt the BBB, interfering with astrocyte endfeet and endothelial tight junction integrity, resulting in altered vascular permeability and an associated CNS inflammation. Aberrant Wnt tone in OPCs mediates their dysfunctional vascular detachment and also leads to OPC secretion of Wif1, which interferes with Wnt ligand function on endothelial tight junction integrity. Evidence for this defective oligodendroglial-vascular interaction in MS suggests that aberrant OPC perivascular migration not only impairs their lesion recruitment but can also act as a disease perpetuator via disruption of the BBB.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Encefalite/fisiopatologia , Esclerose Múltipla/fisiopatologia , Células Precursoras de Oligodendrócitos/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Astrócitos/patologia , Astrócitos/fisiologia , Barreira Hematoencefálica/patologia , Movimento Celular , Células Cultivadas , Encefalite/patologia , Proteínas da Matriz Extracelular/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Esclerose Múltipla/patologia , Células Precursoras de Oligodendrócitos/patologia , Junções Íntimas/metabolismo , Substância Branca/patologia
8.
Glia ; 67(7): 1374-1384, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30861188

RESUMO

It is now well-established that the macrophage and microglial response to CNS demyelination influences remyelination by removing myelin debris and secreting a variety of signaling molecules that influence the behaviour of oligodendrocyte progenitor cells (OPCs). Previous studies have shown that changes in microglia contribute to the age-related decline in the efficiency of remyelination. In this study, we show that microglia increase their expression of the proteoglycan NG2 with age, and that this is associated with an altered micro-niche generated by aged, but not young, microglia that can divert the differentiation OPCs from oligodendrocytes into astrocytes in vitro. We further show that these changes in ageing microglia are generated by exposure to high levels of TGFß. Thus, our findings suggest that the rising levels of circulating TGFß known to occur with ageing contribute to the age-related decline in remyelination by impairing the ability of microglia to promote oligodendrocyte differentiation from OPCs, and therefore could be a potential therapeutic target to promote remyelination.


Assuntos
Senescência Celular/fisiologia , Microglia/metabolismo , Células Precursoras de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fatores Etários , Animais , Animais Recém-Nascidos , Diferenciação Celular/fisiologia , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Relação Dose-Resposta a Droga , Microglia/efeitos dos fármacos , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
9.
Neuron ; 99(4): 689-701.e5, 2018 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-30078577

RESUMO

To address the significance of enhancing myelination for functional recovery after white matter injury (WMI) in preterm infants, we characterized hypomyelination associated with chronic hypoxia and identified structural and functional deficits of excitatory cortical synapses with a prolonged motor deficit. We demonstrate that genetically delaying myelination phenocopies the synaptic and functional deficits observed in mice after hypoxia, suggesting that myelination may possibly facilitate excitatory presynaptic innervation. As a gain-of-function experiment, we specifically ablated the muscarinic receptor 1 (M1R), a negative regulator of oligodendrocyte differentiation in oligodendrocyte precursor cells. Genetically enhancing oligodendrocyte differentiation and myelination rescued the synaptic loss after chronic hypoxia and promoted functional recovery. As a proof of concept, drug-based myelination therapies also resulted in accelerated differentiation and myelination with functional recovery after chronic hypoxia. Together, our data indicate that myelination-enhancing strategies in preterm infants may represent a promising therapeutic approach for structural/functional recovery after hypoxic WMI.


Assuntos
Hipóxia/metabolismo , Bainha de Mielina/fisiologia , Neurogênese/fisiologia , Oligodendroglia/fisiologia , Recuperação de Função Fisiológica/fisiologia , Sinapses/fisiologia , Animais , Animais Recém-Nascidos , Doença Crônica , Feminino , Hipóxia/genética , Hipóxia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Bainha de Mielina/química , Bainha de Mielina/patologia , Receptor Muscarínico M1/deficiência , Sinapses/química , Sinapses/patologia
10.
Nat Commun ; 9(1): 2862, 2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30131568

RESUMO

Oligodendrocyte progenitor cells (OPC) undergo asymmetric cell division (ACD) to generate one OPC and one differentiating oligodendrocyte (OL) progeny. Loss of pro-mitotic proteoglycan and OPC marker NG2 in the OL progeny is the earliest immunophenotypic change of unknown mechanism that indicates differentiation commitment. Here, we report that expression of the mouse homolog of Drosophila tumor suppressor Lethal giant larvae 1 (Lgl1) is induced during OL differentiation. Lgl1 conditional knockout OPC progeny retain NG2 and show reduced OL differentiation, while undergoing more symmetric self-renewing divisions at the expense of asymmetric divisions. Moreover, Lgl1 and hemizygous Ink4a/Arf knockouts in OPC synergistically induce gliomagenesis. Time lapse and total internal reflection microscopy reveals a critical role for Lgl1 in NG2 endocytic routing and links aberrant NG2 recycling to failed differentiation. These data establish Lgl1 as a suppressor of gliomagenesis and positive regulator of asymmetric division and differentiation in the healthy and demyelinated murine brain.


Assuntos
Glicoproteínas/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Proteoglicanas/metabolismo , Animais , Divisão Celular Assimétrica/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Imunofluorescência , Glicoproteínas/genética , Immunoblotting , Camundongos , Monensin/farmacologia , Oligodendroglia/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
11.
Brain ; 141(1): 85-98, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29244098

RESUMO

Hypoxia can injure brain white matter tracts, comprised of axons and myelinating oligodendrocytes, leading to cerebral palsy in neonates and delayed post-hypoxic leukoencephalopathy (DPHL) in adults. In these conditions, white matter injury can be followed by myelin regeneration, but myelination often fails and is a significant contributor to fixed demyelinated lesions, with ensuing permanent neurological injury. Non-myelinating oligodendrocyte precursor cells are often found in lesions in plentiful numbers, but fail to mature, suggesting oligodendrocyte precursor cell differentiation arrest as a critical contributor to failed myelination in hypoxia. We report a case of an adult patient who developed the rare condition DPHL and made a nearly complete recovery in the setting of treatment with clemastine, a widely available antihistamine that in preclinical models promotes oligodendrocyte precursor cell differentiation. This suggested possible therapeutic benefit in the more clinically prevalent hypoxic injury of newborns, and we demonstrate in murine neonatal hypoxic injury that clemastine dramatically promotes oligodendrocyte precursor cell differentiation, myelination, and improves functional recovery. We show that its effect in hypoxia is oligodendroglial specific via an effect on the M1 muscarinic receptor on oligodendrocyte precursor cells. We propose clemastine as a potential therapy for hypoxic brain injuries associated with white matter injury and oligodendrocyte precursor cell maturation arrest.


Assuntos
Clemastina/uso terapêutico , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/etiologia , Antagonistas dos Receptores Histamínicos H1/uso terapêutico , Hipóxia Encefálica/complicações , Recuperação de Função Fisiológica/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Cerebelo/ultraestrutura , Doenças Desmielinizantes/diagnóstico por imagem , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Hipóxia Encefálica/diagnóstico por imagem , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/ultraestrutura , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Nervo Óptico/fisiopatologia , Oxigênio/farmacologia , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/metabolismo
12.
Neuron ; 96(5): 1003-1012.e7, 2017 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-29103804

RESUMO

Blood-brain barrier (BBB) disruption alters the composition of the brain microenvironment by allowing blood proteins into the CNS. However, whether blood-derived molecules serve as extrinsic inhibitors of remyelination is unknown. Here we show that the coagulation factor fibrinogen activates the bone morphogenetic protein (BMP) signaling pathway in oligodendrocyte progenitor cells (OPCs) and suppresses remyelination. Fibrinogen induces phosphorylation of Smad 1/5/8 and inhibits OPC differentiation into myelinating oligodendrocytes (OLs) while promoting an astrocytic fate in vitro. Fibrinogen effects are rescued by BMP type I receptor inhibition using dorsomorphin homolog 1 (DMH1) or CRISPR/Cas9 activin A receptor type I (ACVR1) knockout in OPCs. Fibrinogen and the BMP target Id2 are increased in demyelinated multiple sclerosis (MS) lesions. Therapeutic depletion of fibrinogen decreases BMP signaling and enhances remyelination in vivo. Targeting fibrinogen may be an upstream therapeutic strategy to promote the regenerative potential of CNS progenitors in diseases with remyelination failure.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Fibrinogênio/farmacologia , Células Precursoras de Oligodendrócitos/metabolismo , Remielinização/efeitos dos fármacos , Receptores de Ativinas Tipo I/efeitos dos fármacos , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Fibrinogênio/antagonistas & inibidores , Lisofosfatidilcolinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Bainha de Mielina/metabolismo , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Plasmídeos/genética , Transdução de Sinais/efeitos dos fármacos
13.
Science ; 351(6271): 379-84, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26798014

RESUMO

Oligodendrocytes myelinate axons in the central nervous system and develop from oligodendrocyte precursor cells (OPCs) that must first migrate extensively during brain and spinal cord development. We show that OPCs require the vasculature as a physical substrate for migration. We observed that OPCs of the embryonic mouse brain and spinal cord, as well as the human cortex, emerge from progenitor domains and associate with the abluminal endothelial surface of nearby blood vessels. Migrating OPCs crawl along and jump between vessels. OPC migration in vivo was disrupted in mice with defective vascular architecture but was normal in mice lacking pericytes. Thus, physical interactions with the vascular endothelium are required for OPC migration. We identify Wnt-Cxcr4 (chemokine receptor 4) signaling in regulation of OPC-endothelial interactions and propose that this signaling coordinates OPC migration with differentiation.


Assuntos
Movimento Celular , Córtex Cerebral/embriologia , Células-Tronco Neurais/fisiologia , Neurogênese , Oligodendroglia/fisiologia , Organogênese , Medula Espinal/embriologia , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/embriologia , Córtex Cerebral/irrigação sanguínea , Endotélio Vascular/citologia , Humanos , Camundongos , Células-Tronco Neurais/citologia , Oligodendroglia/citologia , Pericitos/citologia , Pericitos/fisiologia , Receptores CXCR4/metabolismo , Transdução de Sinais , Medula Espinal/irrigação sanguínea , Medula Espinal/citologia , Proteínas Wnt/metabolismo
14.
Dev Neurosci ; 38(6): 430-444, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28343214

RESUMO

Precise temporal and spatial control of the neural stem/progenitor cells within the subventricular zone (SVZ) germinal matrix of the brain is important for normal development in the third trimester and the early postnatal period. The high metabolic demands of proliferating germinal matrix precursors, coupled with the flimsy structure of the germinal matrix cerebral vasculature, are thought to account for the high rates of haemorrhage in extremely- and very-low-birth-weight preterm infants. Germinal matrix haemorrhage can commonly extend to intraventricular haemorrhage (IVH). Because neural stem/progenitor cells are sensitive to microenvironmental cues from the ventricular, intermediate, and basal domains within the germinal matrix, haemorrhage has been postulated to impact neurological outcomes through aberration of normal neural stem/progenitor cell behaviour. We developed an animal model of neonatal germinal matrix haemorrhage using stereotactic injection of autologous blood into the mouse neonatal germinal matrix. Pathological analysis at 4 days postinjury showed high rates of intraventricular extension and ventricular dilatation but low rates of parenchymal disruption outside the germinal zone, recapitulating key features of human "Papile grade III" IVH. At 4 days postinjury we observed proliferation in the wall of the lateral ventricle with significantly increased numbers of transient amplifying cells within the SVZ and the corpus callosum. Analysis at 21 days postinjury revealed that cortical development was also affected, with increased neuronal and concomitant reduced oligodendroglial differentiation. At the molecular level, we showed downregulation of the expression of the transmembrane receptor Notch2 in CD133+ve cells of the SVZ, raising the possibility that the burst of precocious proliferation seen in our experimental mouse model and the skewed differentiation could be mediated by downregulation of the Notch pathway within the proximal/ventricular domain. These findings raise the possibility that Notch regulation plays a critical role in mediating the response of the neonatal SVZ to ischaemic and haemorrhagic insults.


Assuntos
Hemorragia Cerebral/complicações , Ventrículos Laterais/patologia , Células-Tronco Neurais/patologia , Animais , Animais Recém-Nascidos , Divisão Celular/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Receptor Notch2/metabolismo
15.
J Neurosci ; 35(33): 11482-99, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26290228

RESUMO

The Sox family of transcription factors have been widely studied in the context of oligodendrocyte development. However, comparatively little is known about the role of Sox2, especially during CNS remyelination. Here we show that the expression of Sox2 occurs in oligodendrocyte progenitor cells (OPCs) in rodent models during myelination and in activated adult OPCs responding to demyelination, and is also detected in multiple sclerosis lesions. In normal adult white matter of both mice and rats, it is neither expressed by adult OPCs nor by oligodendrocytes (although it is expressed by a subpopulation of adult astrocytes). Overexpression of Sox2 in rat OPCs in vitro maintains the cells in a proliferative state and inhibits differentiation, while Sox2 knockout results in decreased OPC proliferation and survival, suggesting that Sox2 contributes to the expansion of OPCs during the recruitment phase of remyelination. Loss of function in cultured mouse OPCs also results in an impaired ability to undergo normal differentiation in response to differentiation signals, suggesting that Sox2 expression in activated OPCs also primes these cells to eventually undergo differentiation. In vivo studies on remyelination following experimental toxin-induced demyelination in mice with inducible loss of Sox2 revealed impaired remyelination, which was largely due to a profound attenuation of OPC recruitment and likely also due to impaired differentiation. Our results reveal a key role of Sox2 expression in OPCs responding to demyelination, enabling them to effectively contribute to remyelination. SIGNIFICANCE STATEMENT: Understanding the mechanisms of CNS remyelination is central to developing effective means by which this process can be therapeutically enhanced in chronic demyelinating diseases such as multiple sclerosis. In this study, we describe the role of Sox2, a transcription factor widely implicated in stem cell biology, in CNS myelination and remyelination. We show how Sox2 is expressed in oligodendrocyte progenitor cells (OPCs) preparing to undergo differentiation, allowing them to undergo proliferation and priming them for subsequent differentiation. Although Sox2 is unlikely to be a direct therapeutic target, these data nevertheless provide more information on how OPC differentiation is controlled and therefore enriches our understanding of this important CNS regenerative process.


Assuntos
Doenças Desmielinizantes/patologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Fatores de Transcrição SOXB1/metabolismo , Células-Tronco/metabolismo , Células-Tronco/patologia , Animais , Diferenciação Celular , Células Cultivadas , Doenças Desmielinizantes/metabolismo , Feminino , Camundongos , Camundongos Transgênicos , Regeneração Nervosa/fisiologia , Ratos , Ratos Sprague-Dawley
16.
Glia ; 63(10): 1840-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25946682

RESUMO

Wnt signaling plays an essential role in developmental and regenerative myelination of the CNS, therefore it is critical to understand how the factors associated with the various regulatory layers of this complex pathway contribute to these processes. Recently, Apcdd1 was identified as a negative regulator of proximal Wnt signaling, however its role in oligodendrocyte (OL) differentiation and reymelination in the CNS remain undefined. Analysis of Apcdd1 expression revealed dynamic expression during OL development, where its expression is upregulated during differentiation. Functional studies using ex vivo and in vitro OL systems revealed that Apcdd1 promotes OL differentiation, suppresses Wnt signaling, and associates with ß-catenin. Application of these findings to white matter injury (WMI) models revealed that Apcdd1 similarly promotes OL differentiation after gliotoxic injury in vivo and acute hypoxia ex vivo. Examination of Apcdd1 expression in white matter lesions from neonatal WMI and adult multiple sclerosis revealed its expression in subsets of oligodendrocyte (OL) precursors. These studies describe, for the first time, the role of Apcdd1 in OLs after WMI and reveal that negative regulators of the proximal Wnt pathway can influence regenerative myelination, suggesting a new therapeutic strategy for modulating Wnt signaling and stimulating repair after WMI.


Assuntos
Diferenciação Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Oligodendroglia/fisiologia , Doença do Músculo Branco/patologia , Fatores Etários , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Hipóxia/complicações , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lisofosfatidilcolinas/toxicidade , Proteínas de Membrana/genética , Camundongos , Técnicas de Cultura de Órgãos , Medula Espinal/patologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Doença do Músculo Branco/induzido quimicamente , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo
17.
Neuron ; 85(6): 1227-43, 2015 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-25754822

RESUMO

Wnt signaling plays an essential role in developmental and regenerative myelination of the CNS; however, contributions of proximal regulators of the Wnt receptor complex to these processes remain undefined. To identify components of the Wnt pathway that regulate these processes, we applied a multifaceted discovery platform and found that Daam2-PIP5K comprise a novel pathway regulating Wnt signaling and myelination. Using dorsal patterning of the chick spinal cord we found that Daam2 promotes Wnt signaling and receptor complex formation through PIP5K-PIP2. Analysis of Daam2 function in oligodendrocytes (OLs) revealed that it suppresses OL differentiation during development, after white matter injury (WMI), and is expressed in human white matter lesions. These findings suggest a pharmacological strategy to inhibit Daam2-PIP5K function, application of which stimulates remyelination after WMI. Put together, our studies integrate information from multiple systems to identify a novel regulatory pathway for Wnt signaling and potential therapeutic target for WMI.


Assuntos
Proteínas dos Microfilamentos/metabolismo , Oligodendroglia/citologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Medula Espinal/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Diferenciação Celular/fisiologia , Camundongos , Proteínas dos Microfilamentos/genética , Bainha de Mielina/metabolismo , Regeneração/fisiologia , Transdução de Sinais/fisiologia , Medula Espinal/patologia , Substância Branca/lesões , Proteínas rho de Ligação ao GTP/genética
18.
Nat Med ; 20(8): 954-960, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24997607

RESUMO

Functional screening for compounds that promote remyelination represents a major hurdle in the development of rational therapeutics for multiple sclerosis. Screening for remyelination is problematic, as myelination requires the presence of axons. Standard methods do not resolve cell-autonomous effects and are not suited for high-throughput formats. Here we describe a binary indicant for myelination using micropillar arrays (BIMA). Engineered with conical dimensions, micropillars permit resolution of the extent and length of membrane wrapping from a single two-dimensional image. Confocal imaging acquired from the base to the tip of the pillars allows for detection of concentric wrapping observed as 'rings' of myelin. The platform is formatted in 96-well plates, amenable to semiautomated random acquisition and automated detection and quantification. Upon screening 1,000 bioactive molecules, we identified a cluster of antimuscarinic compounds that enhance oligodendrocyte differentiation and remyelination. Our findings demonstrate a new high-throughput screening platform for potential regenerative therapeutics in multiple sclerosis.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Esclerose Múltipla/tratamento farmacológico , Antagonistas Muscarínicos/isolamento & purificação , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Clemastina/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Antagonistas dos Receptores Histamínicos H1/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Antagonistas Muscarínicos/farmacologia , Nanoestruturas , Oligodendroglia/citologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/fisiologia , Ratos , Ratos Sprague-Dawley , Regeneração/efeitos dos fármacos
19.
Cell ; 158(2): 383-396, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25018103

RESUMO

Myelin sheaths provide critical functional and trophic support for axons in white matter tracts of the brain. Oligodendrocyte precursor cells (OPCs) have extraordinary metabolic requirements during development as they differentiate to produce multiple myelin segments, implying that they must first secure adequate access to blood supply. However, mechanisms that coordinate myelination and angiogenesis are unclear. Here, we show that oxygen tension, mediated by OPC-encoded hypoxia-inducible factor (HIF) function, is an essential regulator of postnatal myelination. Constitutive HIF1/2α stabilization resulted in OPC maturation arrest through autocrine activation of canonical Wnt7a/7b. Surprisingly, such OPCs also show paracrine activity that induces excessive postnatal white matter angiogenesis in vivo and directly stimulates endothelial cell proliferation in vitro. Conversely, OPC-specific HIF1/2α loss of function leads to insufficient angiogenesis in corpus callosum and catastrophic axon loss. These findings indicate that OPC-intrinsic HIF signaling couples postnatal white matter angiogenesis, axon integrity, and the onset of myelination in mammalian forebrain.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Animais , Diferenciação Celular , Corpo Caloso/metabolismo , Células Endoteliais/citologia , Técnicas In Vitro , Camundongos , Neovascularização Fisiológica , Células-Tronco Neurais , Oxigênio/metabolismo , Comunicação Parácrina , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Proteínas Wnt/metabolismo
20.
Nat Neurosci ; 17(4): 506-12, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24609463

RESUMO

In colon cancer, mutation of the Wnt repressor APC (encoding adenomatous polyposis coli) leads to a state of aberrant and unrestricted high-activity signaling. However, the relevance of high Wnt tone in non-genetic human disease is unknown. Here we demonstrate that distinct functional states of Wnt activity determine oligodendrocyte precursor cell (OPC) differentiation and myelination. Mouse OPCs with genetic Wnt dysregulation (high tone) express multiple genes in common with colon cancer, including Lef1, Sp5, Ets2, Rnf43 and Dusp4. Surprisingly, we found that OPCs in lesions of hypoxic human neonatal white matter injury upregulated markers of high Wnt activity and lacked expression of APC. We also found that lack of Wnt repressor tone promoted permanent white matter injury after mild hypoxic insult. These findings suggest a state of pathological high-activity Wnt signaling in human disease tissues that lack predisposing genetic mutation.


Assuntos
Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Neoplasias do Colo/fisiopatologia , Hipóxia/metabolismo , Leucoencefalopatias/metabolismo , Oligodendroglia/fisiologia , Proteínas Wnt/fisiologia , Via de Sinalização Wnt/fisiologia , Proteína da Polipose Adenomatosa do Colo/genética , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Biomarcadores/metabolismo , Lesões Encefálicas/patologia , Diferenciação Celular , Neoplasias do Colo/patologia , Feminino , Regulação da Expressão Gênica/fisiologia , Estudos de Associação Genética , Humanos , Recém-Nascido , Doenças do Recém-Nascido , Camundongos , Camundongos Transgênicos , Oligodendroglia/metabolismo , Distribuição Aleatória , Regulação para Cima , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...