Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Dev Cell ; 59(5): 627-644.e10, 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38309265

RESUMO

Axons undergo striking changes in their content and distribution of cell adhesion molecules (CAMs) and ion channels during myelination that underlies the switch from continuous to saltatory conduction. These changes include the removal of a large cohort of uniformly distributed CAMs that mediate initial axon-Schwann cell interactions and their replacement by a subset of CAMs that mediate domain-specific interactions of myelinated fibers. Here, using rodent models, we examine the mechanisms and significance of this removal of axonal CAMs. We show that Schwann cells just prior to myelination locally activate clathrin-mediated endocytosis (CME) in axons, thereby driving clearance of a broad array of axonal CAMs. CAMs engineered to resist endocytosis are persistently expressed along the axon and delay both PNS and CNS myelination. Thus, glia non-autonomously activate CME in axons to downregulate axonal CAMs and presumptively axo-glial adhesion. This promotes the transition from ensheathment to myelination while simultaneously sculpting the formation of axonal domains.


Assuntos
Axônios , Roedores , Humanos , Animais , Axônios/metabolismo , Bainha de Mielina/fisiologia , Células de Schwann , Moléculas de Adesão Celular/metabolismo
2.
bioRxiv ; 2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-38045415

RESUMO

Purpose: The ability of MRI-based markers to detect myelin in the brain is limited. This study investigated the potential of combining multiple MRI markers, each targeting distinct myelin properties, to improve myelin characterization. Methods: We acquired ex vivo multiparametric MRI data at 7 Tesla from control and Gli1 -/- mouse brains at postnatal day 10 (P10), which exhibits enhanced myelination in the corpus callosum, followed by myelin basic protein (MBP) stained immunohistochemistry. Results: Although most MRI markers included in this study showed significant differences in the corpus callosum between control and Gli1 -/- , only fractional anisotropy (FA), mean diffusivity (MD), and T 2 had strong correlations with MBP signals. Partial least square regression (PSLR) based on MRI and MBP values from white matter regions suggested that T 2 had the highest contributions to myelin estimation. When both white and gray matter regions were included, inhomogeneous MT ratio and FA showed strong contributions. Conclusion: This study demonstrates the efficacy of multi-parametric MRI in detecting enhanced myelination in the Gli1 -/- mouse brain. T 2 and diffusion MRI parameters showed strong correlation with MBP signals in the genu of the corpus callosum at P10. The contribution of individual MRI parameter for detecting myelin can be evaluated using PLSR.

3.
Front Cell Neurosci ; 17: 1114781, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36779010

RESUMO

A major therapeutic goal in demyelinating diseases, such as Multiple Sclerosis, is to improve remyelination, thereby restoring effective axon conduction and preventing neurodegeneration. In the adult central nervous system (CNS), parenchymal oligodendrocyte progenitor cells (pOPCs) and, to a lesser extent, pre-existing oligodendrocytes (OLs) and oligodendrocytes generated from neural stem cells (NSCs) in the sub-ventricular zone (SVZ) are capable of forming new myelin sheaths. Due to their self-renewal capabilities and the ability of their progeny to migrate widely within the CNS, NSCs represent an additional source of remyelinating cells that may be targeted to supplement repair by pOPCs. However, in demyelinating disorders and disease models, the NSC contribution to myelin repair is modest and most evident in regions close to the SVZ. We hypothesized that NSC-derived cells may compete with OPCs to remyelinate the same axons, with pOPCs serving as the primary remyelinating cells due to their widespread distribution within the adult CNS, thereby limiting the contribution of NSC-progeny. Here, we have used a dual reporter, genetic fate mapping strategy, to characterize the contribution of pOPCs and NSC-derived OLs to remyelination after cuprizone-induced demyelination. We confirmed that, while pOPCs are the main remyelinating cells in the corpus callosum, NSC-derived cells are also activated and recruited to demyelinating lesions. Blocking pOPC differentiation genetically, resulted in a significant increase in the recruitment NSC-derived cells into the demyelinated corpus callosum and their differentiation into OLs. These results strongly suggest that pOPCs and NSC-progeny compete to repair white matter lesions. They underscore the potential significance of targeting NSCs to improve repair when the contribution of pOPCs is insufficient to affect full remyelination.

4.
J Neurosci ; 42(2): 183-201, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-34772739

RESUMO

Peripheral nerves are organized into discrete compartments. Axons, Schwann cells (SCs), and endoneurial fibroblasts (EFs) reside within the endoneurium and are surrounded by the perineurium, a cellular sheath comprised of layers of perineurial glia (PNG). SC secretion of Desert Hedgehog (Dhh) regulates this organization. In Dhh nulls, the perineurium is deficient and the endoneurium is subdivided into small compartments termed minifascicles. Human Dhh mutations cause a neuropathy with similar defects. Here we examine the role of Gli1, a canonical transcriptional effector of hedgehog signaling, in regulating peripheral nerve organization in mice of both genders. We identify PNG, EFs, and pericytes as Gli1-expressing cells by genetic fate mapping. Although expression of Dhh by SCs and Gli1 in target cells is coordinately regulated with myelination, Gli1 expression unexpectedly persists in Dhh null EFs. Thus, Gli1 is expressed in EFs noncanonically (i.e., independent of hedgehog signaling). Gli1 and Dhh also have nonredundant activities. Unlike Dhh nulls, Gli1 nulls have a normal perineurium. Like Dhh nulls, Gli1 nulls form minifascicles, which we show likely arise from EFs. Thus, Dhh and Gli1 are independent signals: Gli1 is dispensable for perineurial development but functions cooperatively with Dhh to drive normal endoneurial development. During development, Gli1 also regulates endoneurial extracellular matrix production, nerve vascular organization, and has modest, nonautonomous effects on SC sorting and myelination of axons. Finally, in adult nerves, induced deletion of Gli1 is sufficient to drive minifascicle formation. Thus, Gli1 regulates the development and is required to maintain the endoneurial architecture of peripheral nerves.SIGNIFICANCE STATEMENT Peripheral nerves are organized into distinct cellular/ECM compartments: the epineurium, perineurium, and endoneurium. This organization, with its associated cellular constituents, is critical for the structural and metabolic support of nerves and their response to injury. Here, we show that Gli1, a transcription factor normally expressed downstream of hedgehog signaling, is required for the proper organization of the endoneurium but not the perineurium. Unexpectedly, Gli1 expression by endoneurial cells is independent of, and functions nonredundantly with, Schwann Cell-derived Desert Hedgehog in regulating peripheral nerve architecture. These results further delineate how peripheral nerves acquire their distinctive organization during normal development, and highlight mechanisms that may regulate their reorganization in pathologic settings, including peripheral neuropathies and nerve injury.


Assuntos
Nervos Periféricos/metabolismo , Proteína GLI1 em Dedos de Zinco/metabolismo , Animais , Axônios/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/genética , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Células de Schwann/metabolismo , Proteína GLI1 em Dedos de Zinco/genética
5.
Sci Data ; 8(1): 278, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34711861

RESUMO

In the adult mammalian brain, Gli1 expressing neural stem cells reside in the subventricular zone and their progeny are recruited to sites of demyelination in the white matter where they generate new oligodendrocytes, the myelin forming cells. Remarkably, genetic loss or pharmacologic inhibition of Gli1 enhances the efficacy of remyelination by these neural stem cells. To understand the molecular mechanisms involved, we performed a transcriptomic analysis of this Gli1-pool of neural stem cells. We compared murine NSCs with either intact or deficient Gli1 expression from adult mice on a control diet or on a cuprizone diet which induces widespread demyelination. These data will be a valuable resource for identifying therapeutic targets for enhancing remyelination in demyelinating diseases like multiple sclerosis.


Assuntos
Doenças Desmielinizantes/genética , Células-Tronco Neurais/citologia , Transcriptoma , Proteína GLI1 em Dedos de Zinco/genética , Animais , Cuprizona , Camundongos , Oligodendroglia/citologia
6.
Bio Protoc ; 11(12): e4067, 2021 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-34263008

RESUMO

The function of neurons in afferent reception, integration, and generation of electrical activity relies on their strikingly polarized organization, characterized by distinct membrane domains. These domains have different compositions resulting from a combination of selective targeting and retention of membrane proteins. In neurons, most proteins are delivered from their site of synthesis in the soma to the axon via anterograde vesicular transport and undergo retrograde transport for redistribution and/or lysosomal degradation. A key question is whether proteins destined for the same domain are transported in separate vesicles for local assembly or whether these proteins are pre-assembled and co-transported in the same vesicles for delivery to their cognate domains. To assess the content of transport vesicles, one strategy relies on staining of sciatic nerves after ligation, which drives the accumulation of anterogradely and retrogradely transported vesicles on the proximal and distal side of the ligature, respectively. This approach may not permit confident assessment of the nature of the intracellular vesicles identified by staining, and analysis is limited to the availability of suitable antibodies. Here, we use dual color live imaging of proteins labeled with different fluorescent tags, visualizing anterograde and retrograde axonal transport of several proteins simultaneously. These proteins were expressed in rat dorsal root ganglion (DRG) neurons cultured alone or with Schwann cells under myelinating conditions to assess whether glial cells modify the patterns of axonal transport. Advantages of this protocol are the dynamic identification of transport vesicles and characterization of their content for various proteins that is not limited by available antibodies.

7.
Glia ; 69(6): 1583-1604, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33620118

RESUMO

Microgliosis is a prominent pathological feature in many neurological diseases including multiple sclerosis (MS), a progressive auto-immune demyelinating disorder. The precise role of microglia, parenchymal central nervous system (CNS) macrophages, during demyelination, and the relative contributions of peripheral macrophages are incompletely understood. Classical markers used to identify microglia do not reliably discriminate between microglia and peripheral macrophages, confounding analyses. Here, we use a genetic fate mapping strategy to identify microglia as predominant responders and key effectors of demyelination in the cuprizone (CUP) model. Colony-stimulating factor 1 (CSF1), also known as macrophage colony-stimulating factor (M-CSF) - a secreted cytokine that regulates microglia development and survival-is upregulated in demyelinated white matter lesions. Depletion of microglia with the CSF1R inhibitor PLX3397 greatly abrogates the demyelination, loss of oligodendrocytes, and reactive astrocytosis that results from CUP treatment. Electron microscopy (EM) and serial block face imaging show myelin sheaths remain intact in CUP treated mice depleted of microglia. However, these CUP-damaged myelin sheaths are lost and robustly phagocytosed upon-repopulation of microglia. Direct injection of CSF1 into CNS white matter induces focal microgliosis and demyelination indicating active CSF1 signaling can promote demyelination. Finally, mice defective in adopting a toxic astrocyte phenotype that is driven by microglia nevertheless demyelinate normally upon CUP treatment implicating microglia rather than astrocytes as the primary drivers of CUP-mediated demyelination. Together, these studies indicate activated microglia are required for and can drive demyelination directly and implicate CSF1 signaling in these events.


Assuntos
Doenças Desmielinizantes , Microglia , Animais , Cuprizona/toxicidade , Doenças Desmielinizantes/induzido quimicamente , Modelos Animais de Doenças , Macrófagos , Camundongos , Receptores de Fator Estimulador de Colônias , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Transdução de Sinais
8.
Stem Cell Reports ; 15(5): 1047-1055, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33125874

RESUMO

Enhancing repair of myelin is an important therapeutic goal in many neurological disorders characterized by demyelination. In the healthy adult brain, ventral neural stem cells (vNSCs) in the subventricular zone, marked by GLI1 expression, do not generate oligodendrocytes. However, in response to demyelination, their progeny are recruited to lesions where they differentiate into oligodendrocytes and ablation of GLI1 further enhances remyelination. GLI1 and GLI2 are closely related transcriptional activators but the role of GLI2 in remyelination by vNSCs is not clear. Here, we show that genetic ablation of Gli1 in vNSCs increases GLI2 expression and combined loss of both transcription factors decreases the recruitment and differentiation of their progeny in demyelinated lesions. These results indicate that GLI1 and GLI2 have distinct, non-redundant functions in vNSCs and their relative levels play an essential role in the response to demyelination.


Assuntos
Doenças Desmielinizantes/metabolismo , Células-Tronco Neurais/metabolismo , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína Gli2 com Dedos de Zinco/metabolismo , Animais , Diferenciação Celular , Doenças Desmielinizantes/genética , Ventrículos Laterais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Remielinização , Deleção de Sequência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima , Proteína GLI1 em Dedos de Zinco/genética , Proteína Gli2 com Dedos de Zinco/genética
9.
J Neurosci ; 40(30): 5709-5723, 2020 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-32554548

RESUMO

The paranodal junctions flank mature nodes of Ranvier and provide a barrier between ion channels at the nodes and juxtaparanodes. These junctions also promote node assembly and maintenance by mechanisms that are poorly understood. Here, we examine their role in the accumulation of NF186, a key adhesion molecule of PNS and CNS nodes. We previously showed that NF186 is initially targeted/accumulates via its ectodomain to forming PNS (hemi)nodes by diffusion trapping, whereas it is later targeted to mature nodes by a transport-dependent mechanism mediated by its cytoplasmic segment. To address the role of the paranodes in this switch, we compared accumulation of NF186 ectodomain and cytoplasmic domain constructs in WT versus paranode defective (i.e., Caspr-null) mice. Both pathways are affected in the paranodal mutants. In the PNS of Caspr-null mice, diffusion trapping mediated by the NF186 ectodomain aberrantly persists into adulthood, whereas the cytoplasmic domain/transport-dependent targeting is impaired. In contrast, accumulation of NF186 at CNS nodes does not undergo a switch; it is predominantly targeted to both forming and mature CNS nodes via its cytoplasmic domain and requires intact paranodes. Fluorescence recovery after photobleaching analysis indicates that the paranodes provide a membrane diffusion barrier that normally precludes diffusion of NF186 to nodes. Linkage of paranodal proteins to the underlying cytoskeleton likely contributes to this diffusion barrier based on 4.1B and ßII spectrin expression in Caspr-null mice. Together, these results implicate the paranodes as membrane diffusion barriers that regulate targeting to nodes and highlight differences in the assembly of PNS and CNS nodes.SIGNIFICANCE STATEMENT Nodes of Ranvier are essential for effective saltatory conduction along myelinated axons. A major question is how the various axonal proteins that comprise the multimeric nodal complex accumulate at this site. Here we examine how targeting of NF186, a key nodal adhesion molecule, is regulated by the flanking paranodal junctions. We show that the transition from diffusion-trapping to transport-dependent accumulation of NF186 requires the paranodal junctions. We also demonstrate that these junctions are a barrier to diffusion of axonal proteins into the node and highlight differences in PNS and CNS node assembly. These results provide new insights into the mechanism of node assembly and the pathophysiology of neurologic disorders in which impaired paranodal function contributes to clinical disability.


Assuntos
Moléculas de Adesão Celular/metabolismo , Gânglios Espinais/metabolismo , Fatores de Crescimento Neural/metabolismo , Nós Neurofibrosos/metabolismo , Animais , Moléculas de Adesão Celular/análise , Células Cultivadas , Feminino , Gânglios Espinais/química , Gânglios Espinais/citologia , Junções Intercelulares/química , Junções Intercelulares/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fatores de Crescimento Neural/análise , Nós Neurofibrosos/química
10.
Neuron ; 106(5): 707-709, 2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32497505

RESUMO

Enrichment of sodium channels at nodes of Ranvier, a hallmark of myelinated axons, underlies effective saltatory conduction. In this issue of Neuron, Eshed-Eisenbach et al. (2020) demonstrate that proteolysis of gliomedin, which drives initial channel clustering, provides a novel mechanism to ensure fidelity of channel localization to nodes.


Assuntos
Peptídeo Hidrolases , Nós Neurofibrosos , Axônios , Proteína Morfogenética Óssea 1 , Análise por Conglomerados
11.
J Cell Biol ; 219(6)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32289157

RESUMO

Neurons are highly polarized cells organized into functionally and molecularly distinct domains. A key question is whether the multiprotein complexes that comprise these domains are preassembled, transported, and inserted as a complex or whether their components are transported independently and assemble locally. Here, we have dynamically imaged, in pairwise combinations, the vesicular transport of fluorescently tagged components of the nodes of Ranvier and other myelinated axonal domains in sensory neurons cultured alone or together with Schwann cells at the onset of myelination. In general, most proteins are transported independently in the anterograde direction. In contrast, there is substantial cotransport of proteins from distinct domains in the retrograde direction likely due to coendocytosis along the axon. Early myelination did not substantially change these patterns of transport, although it increased the overall numbers of axonal transport vesicles. Our results indicate domain components are transported in separate vesicles for local assembly, not as preformed complexes, and implicate endocytosis along axons as a mechanism of clearance.


Assuntos
Transporte Axonal/fisiologia , Axônios/metabolismo , Endocitose/genética , Bainha de Mielina/metabolismo , Vesículas Transportadoras/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animais , Anquirinas/genética , Anquirinas/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Endocitose/fisiologia , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Neurônios/metabolismo , Domínios Proteicos , Ratos , Ratos Sprague-Dawley , Células de Schwann , Proteínas de Transporte Vesicular/genética
13.
Glia ; 67(5): 884-895, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30585357

RESUMO

Interactions between axons and Schwann cells are essential for the acquisition of Schwann cell radial and longitudinal polarity and myelin sheath assembly. In the internode, the largest of these longitudinal domains, axon-Schwann cell interactions are mediated by the Nectin-like (Necl) cell adhesion proteins, also known as SynCAMs or Cadms. In particular, Necl-1/Cadm3 expressed on the axon surface binds to Necl-4/Cadm4 expressed along the adaxonal membrane of myelinating Schwann cells. Necl-4 promotes myelination in vitro and is required for the timely onset of myelination and the fidelity of the organization of the myelin sheath and the internode in vivo. A key question is the identity of the downstream effectors of Necl-4 that mediate its effects. The cytoplasmic terminal region (CTR) of Necl-4 contains a PDZ-domain binding motif. Accordingly, we used the CTR of Necl-4 in an unbiased proteomic screen of PDZ-domain proteins. We identify Par-3, a multi-PDZ domain containing protein of the Par-aPKC polarity complex previously implicated in myelination, as an interacting protein. Necl-4 and Par-3 are colocalized along the inner Schwann cell membrane and coprecipitate from Schwann cell lysates. The CTR of Necl-4 binds to the first PDZ domain of Par-3 thereby recruiting Par-3 to sites of Necl-4/Necl-1 interaction. Knockdown of Necl-4 perturbs Par-3 localization to the inner membrane of Schwann cells in myelinating co-cultures. These findings implicate interactions of Necl-1/Necl-4 in the recruitment of Par-3 to the Schwann cell adaxonal membrane and the establishment of Schwann cell radial polarity.


Assuntos
Axônios/metabolismo , Moléculas de Adesão Celular/metabolismo , Membrana Celular/metabolismo , Imunoglobulinas/metabolismo , Domínios PDZ/fisiologia , Células de Schwann/citologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Células CHO , Moléculas de Adesão Celular/genética , Proteínas de Ciclo Celular , Membrana Celular/genética , Técnicas de Cocultura , Cricetulus , Embrião de Mamíferos , Gânglios Espinais/citologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Imunoglobulinas/genética , Imunoprecipitação , Técnicas In Vitro , Camundongos , Neurônios , Domínios PDZ/genética , Ratos , Nervo Isquiático/citologia , Transfecção
14.
Neuron ; 97(3): 555-570.e6, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29395909

RESUMO

The axon initial segment (AIS) is the site of action potential generation and a locus of activity-dependent homeostatic plasticity. A multimeric complex of sodium channels, linked via a cytoskeletal scaffold of ankyrin G and beta IV spectrin to submembranous actin rings, mediates these functions. The mechanisms that specify the AIS complex to the proximal axon and underlie its plasticity remain poorly understood. Here we show phosphorylated myosin light chain (pMLC), an activator of contractile myosin II, is highly enriched in the assembling and mature AIS, where it associates with actin rings. MLC phosphorylation and myosin II contractile activity are required for AIS assembly, and they regulate the distribution of AIS components along the axon. pMLC is rapidly lost during depolarization, destabilizing actin and thereby providing a mechanism for activity-dependent structural plasticity of the AIS. Together, these results identify pMLC/myosin II activity as a common link between AIS assembly and plasticity.


Assuntos
Actinas/metabolismo , Segmento Inicial do Axônio/metabolismo , Cadeias Leves de Miosina/metabolismo , Miosina Tipo II/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Córtex Cerebral/metabolismo , Feminino , Hipocampo/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatase de Miosina-de-Cadeia-Leve/genética , Fosforilação , Cultura Primária de Células , Ratos Sprague-Dawley
15.
J Neurosci ; 36(16): 4506-21, 2016 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-27098694

RESUMO

The signaling pathways that regulate myelination in the PNS remain poorly understood. Phosphatidylinositol-4,5-bisphosphate 3-kinase 1A, activated in Schwann cells by neuregulin and the extracellular matrix, has an essential role in the early events of myelination. Akt/PKB, a key effector of phosphatidylinositol-4,5-bisphosphate 3-kinase 1A, was previously implicated in CNS, but not PNS myelination. Here we demonstrate that Akt plays a crucial role in axon ensheathment and in the regulation of myelin sheath thickness in the PNS. Pharmacological inhibition of Akt in DRG neuron-Schwann cell cocultures dramatically decreased MBP and P0 levels and myelin sheath formation without affecting expression of Krox20/Egr2, a key transcriptional regulator of myelination. Conversely, expression of an activated form of Akt in purified Schwann cells increased expression of myelin proteins, but not Krox20/Egr2, and the levels of activated Rac1. Transgenic mice expressing a membrane-targeted, activated form of Akt under control of the 2',3'-cyclic nucleotide 3'-phosphodiesterase promoter, exhibited thicker PNS and CNS myelin sheaths, and PNS myelin abnormalities, such as tomacula and myelin infoldings/outfoldings, centered around the paranodes and Schmidt Lanterman incisures. These effects were corrected by rapamycin treatmentin vivo Importantly, Akt activity in the transgenic mice did not induce myelination of nonmyelinating Schwann cells in the sympathetic trunk or Remak fibers of the dorsal roots, although, in those structures, they wrapped membranes redundantly around axons. Together, our data indicate that Akt is crucial for PNS myelination driving axonal wrapping by unmyelinated and myelinated Schwann cells and enhancing myelin protein synthesis in myelinating Schwann cells. SIGNIFICANCE STATEMENT: Although the role of the key serine/threonine kinase Akt in promoting CNS myelination has been demonstrated, its role in the PNS has not been established and remains uncertain. This work reveals that Akt controls several key steps of the PNS myelination. First, its activity promotes membrane production and axonal wrapping independent of a transcriptional effect. In myelinated axons, it also enhances myelin thickness through the mTOR pathway. Finally, sustained Akt activation in Schwann cells leads to hypermyelination/dysmyelination, mimicking some features present in neuropathies, such as hereditary neuropathy with liability to pressure palsies or demyelinating forms of Charcot-Marie-Tooth disease. Together, these data demonstrate the role of Akt in regulatory mechanisms underlying axonal wrapping and myelination in the PNS.


Assuntos
Axônios/fisiologia , Bainha de Mielina/fisiologia , Proteína Oncogênica v-akt/fisiologia , Nervo Isquiático/fisiologia , Animais , Axônios/ultraestrutura , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Bainha de Mielina/ultraestrutura , Nervos Periféricos/fisiologia , Nervos Periféricos/ultraestrutura , Sistema Nervoso Periférico/fisiologia , Sistema Nervoso Periférico/ultraestrutura , Nervo Isquiático/ultraestrutura
16.
Nature ; 526(7573): 448-52, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26416758

RESUMO

Enhancing repair of myelin is an important but still elusive therapeutic goal in many neurological disorders. In multiple sclerosis, an inflammatory demyelinating disease, endogenous remyelination does occur but is frequently insufficient to restore function. Both parenchymal oligodendrocyte progenitor cells and endogenous adult neural stem cells resident within the subventricular zone are known sources of remyelinating cells. Here we characterize the contribution to remyelination of a subset of adult neural stem cells, identified by their expression of Gli1, a transcriptional effector of the sonic hedgehog pathway. We show that these cells are recruited from the subventricular zone to populate demyelinated lesions in the forebrain but never enter healthy, white matter tracts. Unexpectedly, recruitment of this pool of neural stem cells, and their differentiation into oligodendrocytes, is significantly enhanced by genetic or pharmacological inhibition of Gli1. Importantly, complete inhibition of canonical hedgehog signalling was ineffective, indicating that the role of Gli1 both in augmenting hedgehog signalling and in retarding myelination is specialized. Indeed, inhibition of Gli1 improves the functional outcome in a relapsing/remitting model of experimental autoimmune encephalomyelitis and is neuroprotective. Thus, endogenous neural stem cells can be mobilized for the repair of demyelinated lesions by inhibiting Gli1, identifying a new therapeutic avenue for the treatment of demyelinating disorders.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Bainha de Mielina/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Substância Branca/metabolismo , Substância Branca/patologia , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Diferenciação Celular , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Feminino , Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Ventrículos Laterais , Camundongos , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Fármacos Neuroprotetores/antagonistas & inibidores , Fármacos Neuroprotetores/metabolismo , Oligodendroglia/citologia , Prosencéfalo/metabolismo , Prosencéfalo/patologia , Transdução de Sinais , Substância Branca/citologia , Proteína GLI1 em Dedos de Zinco
17.
Dev Cell ; 34(2): 129-30, 2015 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-26218317

RESUMO

The mechanisms that drive the spiral wrapping of the myelin sheath around axons are poorly understood. Two papers in this issue of Developmental Cell demonstrate that actin disassembly, rather than actin assembly, predominates during oligodendrocyte maturation and is critical for the genesis of the central myelin sheath.


Assuntos
Citoesqueleto de Actina/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Sistema Nervoso Central/crescimento & desenvolvimento , Bainha de Mielina/fisiologia , Oligodendroglia/fisiologia , Animais
18.
Cold Spring Harb Perspect Biol ; 7(8): a020529, 2015 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-26054742

RESUMO

Myelinated nerve fibers are essential for the rapid propagation of action potentials by saltatory conduction. They form as the result of reciprocal interactions between axons and Schwann cells. Extrinsic signals from the axon, and the extracellular matrix, drive Schwann cells to adopt a myelinating fate, whereas myelination reorganizes the axon for its role in conduction and is essential for its integrity. Here, we review our current understanding of the development, molecular organization, and function of myelinating Schwann cells. Recent findings into the extrinsic signals that drive Schwann cell myelination, their cognate receptors, and the downstream intracellular signaling pathways they activate will be described. Together, these studies provide important new insights into how these pathways converge to activate the transcriptional cascade of myelination and remodel the actin cytoskeleton that is critical for morphogenesis of the myelin sheath.


Assuntos
Bainha de Mielina/fisiologia , Fibras Nervosas Mielinizadas/fisiologia , Células de Schwann/fisiologia , Potenciais de Ação , Epigênese Genética , Humanos , Transdução de Sinais , Transcrição Gênica
19.
Proc Natl Acad Sci U S A ; 111(50): 18025-30, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25453108

RESUMO

Understanding the dynamic axon-glial cell interaction underlying myelination is hampered by the lack of suitable imaging techniques. Here we demonstrate third harmonic generation microscopy (THGM) for label-free imaging of myelinating Schwann cells in live culture and ex vivo and in vivo tissue. A 3D structure was acquired for a variety of compact and noncompact myelin domains, including juxtaparanodes, Schmidt-Lanterman incisures, and Cajal bands. Other subcellular features of Schwann cells that escape traditional optical microscopies were also visualized. We tested THGM for morphometry of compact myelin. Unlike current methods based on electron microscopy, g-ratio could be determined along an extended length of myelinated fiber in the physiological condition. The precision of THGM-based g-ratio estimation was corroborated in mouse models of hypomyelination. Finally, we demonstrated the feasibility of THGM to monitor morphological changes of myelin during postnatal development and degeneration. The outstanding capabilities of THGM may be useful for elucidation of the mechanism of myelin formation and pathogenesis.


Assuntos
Imageamento Tridimensional/métodos , Microscopia/métodos , Bainha de Mielina/química , Células de Schwann/citologia , Animais , Doenças Desmielinizantes/patologia , Lasers , Camundongos , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Ratos
20.
J Cell Biol ; 204(7): 1219-36, 2014 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-24687281

RESUMO

The PI 3-kinase (PI 3-K) signaling pathway is essential for Schwann cell myelination. Here we have characterized PI 3-K effectors activated during myelination by probing myelinating cultures and developing nerves with an antibody that recognizes phosphorylated substrates for this pathway. We identified a discrete number of phospho-proteins including the S6 ribosomal protein (S6rp), which is down-regulated at the onset of myelination, and N-myc downstream-regulated gene-1 (NDRG1), which is up-regulated strikingly with myelination. We show that type III Neuregulin1 on the axon is the primary activator of S6rp, an effector of mTORC1. In contrast, laminin-2 in the extracellular matrix (ECM), signaling through the α6ß4 integrin and Sgk1 (serum and glucocorticoid-induced kinase 1), drives phosphorylation of NDRG1 in the Cajal bands of the abaxonal compartment. Unexpectedly, mice deficient in α6ß4 integrin signaling or Sgk1 exhibit hypermyelination during development. These results identify functionally and spatially distinct PI 3-K pathways: an early, pro-myelinating pathway driven by axonal Neuregulin1 and a later-acting, laminin-integrin-dependent pathway that negatively regulates myelination.


Assuntos
Bainha de Mielina/fisiologia , Sistema Nervoso Periférico/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Técnicas de Cocultura , Matriz Extracelular/metabolismo , Expressão Gênica , Proteínas Imediatamente Precoces/metabolismo , Integrina beta4/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Neuregulina-1/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptores de Laminina/metabolismo , Proteína S6 Ribossômica/metabolismo , Células de Schwann/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...