Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
8.
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
9.
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
10.
J Biol Chem ; 287(46): 39061-9, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22992729

RESUMO

Voltage-gated Na(+) channels in the brain are composed of a single pore-forming α subunit, one non-covalently linked ß subunit (ß1 or ß3), and one disulfide-linked ß subunit (ß2 or ß4). The final step in Na(+) channel biosynthesis in central neurons is concomitant α-ß2 disulfide linkage and insertion into the plasma membrane. Consistent with this, Scn2b (encoding ß2) null mice have reduced Na(+) channel cell surface expression in neurons, and action potential conduction is compromised. Here we generated a series of mutant ß2 cDNA constructs to investigate the cysteine residue(s) responsible for α-ß2 subunit covalent linkage. We demonstrate that a single cysteine-to-alanine substitution at extracellular residue Cys-26, located within the immunoglobulin (Ig) domain, abolishes the covalent linkage between α and ß2 subunits. Loss of α-ß2 covalent complex formation disrupts the targeting of ß2 to nodes of Ranvier in a myelinating co-culture system and to the axon initial segment in primary hippocampal neurons, suggesting that linkage with α is required for normal ß2 subcellular localization in vivo. WT ß2 subunits are resistant to live cell Triton X-100 detergent extraction from the hippocampal axon initial segment, whereas mutant ß2 subunits, which cannot form disulfide bonds with α, are removed by detergent. Taken together, our results demonstrate that α-ß2 covalent association via a single, extracellular disulfide bond is required for ß2 targeting to specialized neuronal subcellular domains and for ß2 association with the neuronal cytoskeleton within those domains.


Assuntos
Cisteína/química , Canal de Sódio Disparado por Voltagem NAV1.1/química , Animais , Encéfalo/metabolismo , Adesão Celular , Membrana Celular/metabolismo , Técnicas de Cocultura , Citoesqueleto/metabolismo , Dissulfetos/química , Epitopos/química , Células HEK293 , Hipocampo/metabolismo , Humanos , Imuno-Histoquímica/métodos , Mutação , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Neurônios/metabolismo , Mapeamento de Interação de Proteínas/métodos , Ratos , Células de Schwann , Canais de Sódio/química
11.
Glia ; 61(2): 240-53, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23109359

RESUMO

Myelinated axons are organized into specialized domains critical to their function in saltatory conduction, i.e., nodes, paranodes, juxtaparanodes, and internodes. Here, we describe the distribution and role of the 4.1B protein in this organization. 4.1B is expressed by neurons, and at lower levels by Schwann cells, which also robustly express 4.1G. Immunofluorescence and immuno-EM demonstrates 4.1B is expressed subjacent to the axon membrane in all domains except the nodes. Mice deficient in 4.1B have preserved paranodes, based on marker staining and EM in contrast to the juxtaparanodes, which are substantially affected in both the PNS and CNS. The juxtaparanodal defect is evident in developing and adult nerves and is neuron-autonomous based on myelinating cocultures in which wt Schwann cells were grown with 4.1B-deficient neurons. Despite the juxtaparanodal defect, nerve conduction velocity is unaffected. Preservation of paranodal markers in 4.1B deficient mice is associated with, but not dependent on an increase of 4.1R at the axonal paranodes. Loss of 4.1B in the axon is also associated with reduced levels of the internodal proteins, Necl-1 and Necl-2, and of alpha-2 spectrin. Mutant nerves are modestly hypermyelinated and have increased numbers of Schmidt-Lanterman incisures, increased expression of 4.1G, and express a residual, truncated isoform of 4.1B. These results demonstrate that 4.1B is a key cytoskeletal scaffold for axonal adhesion molecules expressed in the juxtaparanodal and internodal domains that unexpectedly regulates myelin sheath thickness.


Assuntos
Proteínas dos Microfilamentos/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Neurônios/citologia , Células de Schwann/metabolismo , Animais , Anquirinas/metabolismo , Axônios/metabolismo , Axônios/ultraestrutura , Molécula 1 de Adesão Celular , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Estimulação Elétrica , Embrião de Mamíferos , Comportamento Exploratório/fisiologia , Gânglios Espinais/citologia , Imunoglobulinas/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Microscopia Eletrônica de Transmissão , Microscopia Imunoeletrônica , Proteína Básica da Mielina/metabolismo , Proteína P0 da Mielina/metabolismo , Proteínas da Mielina/metabolismo , Condução Nervosa/genética , Condução Nervosa/fisiologia , Nós Neurofibrosos/metabolismo , Nós Neurofibrosos/ultraestrutura , Células de Schwann/ultraestrutura , Espectrina/metabolismo
12.
J Cell Sci ; 124(Pt 22): 3784-96, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22100921

RESUMO

Signaling through cyclic AMP (cAMP) has been implicated in the regulation of Schwann cell (SC) proliferation and differentiation. In quiescent SCs, elevation of cAMP promotes the expression of proteins associated with myelination such as Krox-20 and P0, and downregulation of markers associated with the non-myelinating SC phenotype. We have previously shown that the motor protein myosin II is required for the establishment of normal SC-axon interactions, differentiation and myelination, however, the mechanisms behind these effects are unknown. Here we report that the levels and activity of myosin light chain kinase (MLCK), an enzyme that regulates MLC phosphorylation in non-muscle cells, are dramatically downregulated in SCs after cAMP treatment, in a similar pattern to that of c-Jun, a known inhibitor of myelination. Knockdown of MLCK in SCs mimics the effect of cAMP elevation, inducing plasma membrane expansion and expression of Krox-20 and myelin proteins. Despite activation of myelin gene transcription these cells fail to make compact myelin when placed in contact with axons. Our data indicate that myosin II activity is differentially regulated at various stages during myelination and that in the absence of MLCK the processes of SC differentiation and compact myelin assembly are uncoupled.


Assuntos
Diferenciação Celular , Citoesqueleto/metabolismo , Bainha de Mielina/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Células de Schwann/citologia , Células de Schwann/enzimologia , Animais , Células Cultivadas , Quinase de Cadeia Leve de Miosina/genética , Ratos , Células de Schwann/metabolismo
13.
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.

14.
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.

15.
J Neurosci ; 31(34): 12208-17, 2011 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-21865464

RESUMO

Myelinating glial cells exhibit a spectacular cytoarchitecture, because they polarize on multiple axes and domains. How this occurs is essentially unknown. The dystroglycan-dystrophin complex is required for the function of myelin-forming Schwann cells. Similar to other tissues, the dystroglycan complex in Schwann cells localizes with different dystrophin family members in specific domains, thus promoting polarization. We show here that cleavage of dystroglycan by matrix metalloproteinases 2 and 9, an event that is considered pathological in most tissues, is finely and dynamically regulated in normal nerves and modulates dystroglycan complex composition and the size of Schwann cell compartments. In contrast, in nerves of Dy(2j/2j) mice, a model of laminin 211 deficiency, metalloproteinases 2 and 9 are increased, causing excessive dystroglycan cleavage and abnormal compartments. Pharmacological inhibition of cleavage rescues the cytoplasmic defects of Dy(2j/2j) Schwann cells. Thus, regulated cleavage may be a general mechanism to regulate protein complex composition in physiological conditions, whereas unregulated processing is pathogenic and a target for treatment in disease.


Assuntos
Compartimento Celular/fisiologia , Distroglicanas/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Bainha de Mielina/metabolismo , Domínios e Motivos de Interação entre Proteínas/fisiologia , Células de Schwann/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Distroglicanas/química , Metaloproteinase 2 da Matriz/química , Metaloproteinase 2 da Matriz/fisiologia , Metaloproteinase 9 da Matriz/química , Metaloproteinase 9 da Matriz/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Bainha de Mielina/enzimologia , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Ratos , Células de Schwann/enzimologia , Nervo Isquiático/química , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia
16.
J Cell Biol ; 177(5): 857-70, 2007 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-17548513

RESUMO

Axon initial segments (AISs) and nodes of Ranvier are sites of action potential generation and propagation, respectively. Both domains are enriched in sodium channels complexed with adhesion molecules (neurofascin [NF] 186 and NrCAM) and cytoskeletal proteins (ankyrin G and betaIV spectrin). We show that the AIS and peripheral nervous system (PNS) nodes both require ankyrin G but assemble by distinct mechanisms. The AIS is intrinsically specified; it forms independent of NF186, which is targeted to this site via intracellular interactions that require ankyrin G. In contrast, NF186 is targeted to the node, and independently cleared from the internode, by interactions of its ectodomain with myelinating Schwann cells. NF186 is critical for and initiates PNS node assembly by recruiting ankyrin G, which is required for the localization of sodium channels and the entire nodal complex. Thus, initial segments assemble from the inside out driven by the intrinsic accumulation of ankyrin G, whereas PNS nodes assemble from the outside in, specified by Schwann cells, which direct the NF186-dependent recruitment of ankyrin G.


Assuntos
Anquirinas/fisiologia , Axônios/metabolismo , Nós Neurofibrosos/metabolismo , Animais , Anquirinas/antagonistas & inibidores , Axônios/fisiologia , Axônios/ultraestrutura , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Fatores de Crescimento Neural/química , Fatores de Crescimento Neural/metabolismo , Estrutura Terciária de Proteína , Nós Neurofibrosos/fisiologia , Nós Neurofibrosos/ultraestrutura , Ratos , Canais de Sódio/metabolismo
17.
J Cell Biol ; 178(5): 861-74, 2007 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-17724124

RESUMO

Axon-glial interactions are critical for the induction of myelination and the domain organization of myelinated fibers. Although molecular complexes that mediate these interactions in the nodal region are known, their counterparts along the internode are poorly defined. We report that neurons and Schwann cells express distinct sets of nectin-like (Necl) proteins: axons highly express Necl-1 and -2, whereas Schwann cells express Necl-4 and lower amounts of Necl-2. These proteins are strikingly localized to the internode, where Necl-1 and -2 on the axon are directly apposed by Necl-4 on the Schwann cell; all three proteins are also enriched at Schmidt-Lanterman incisures. Binding experiments demonstrate that the Necl proteins preferentially mediate heterophilic rather than homophilic interactions. In particular, Necl-1 on axons binds specifically to Necl-4 on Schwann cells. Knockdown of Necl-4 by short hairpin RNA inhibits Schwann cell differentiation and subsequent myelination in cocultures. These results demonstrate a key role for Necl-4 in initiating peripheral nervous system myelination and implicate the Necl proteins as mediators of axo-glial interactions along the internode.


Assuntos
Axônios/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Isoformas de Proteínas/metabolismo , Nós Neurofibrosos , Células de Schwann/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Axônios/ultraestrutura , Células CHO , Adesão Celular/fisiologia , Moléculas de Adesão Celular , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Cricetinae , Cricetulus , Gânglios Espinais/metabolismo , Imunoglobulinas , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/metabolismo , Bainha de Mielina/ultraestrutura , Fibras Nervosas Mielinizadas/ultraestrutura , Isoformas de Proteínas/genética , Interferência de RNA , Nós Neurofibrosos/metabolismo , Nós Neurofibrosos/ultraestrutura , Ratos , Células de Schwann/citologia , Nervo Isquiático/citologia , Nervo Isquiático/metabolismo , Proteínas Supressoras de Tumor/genética
18.
J Peripher Nerv Syst ; 17 Suppl 3: 14-9, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23279426

RESUMO

Axons in the vertebrate peripheral nervous system are intimately associated with Schwann cells. Axons regulate the Schwann cell phenotype, determining whether they myelinate individual axons or ensheathe multiple, small axons in Remak bundles. Our current understanding of the axonal signals that drive Schwann cells towards these distinct morphological and phenotypic fates is briefly reviewed here. Elucidation of these signals, and the intracellular pathways they regulate, may lead to new, rational therapies for the treatment of inherited and acquired neuropathies.


Assuntos
Axônios/fisiologia , Bainha de Mielina/fisiologia , Células de Schwann/fisiologia , Animais , Comunicação Celular/fisiologia , Humanos , Transdução de Sinais/fisiologia
19.
J Neurosci ; 30(17): 6122-31, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20427670

RESUMO

Members of the neuregulin-1 (Nrg1) growth factor family play important roles during Schwann cell development. Recently, it has been shown that the membrane-bound type III isoform is required for Schwann cell myelination. Interestingly, however, Nrg1 type II, a soluble isoform, inhibits the process. The mechanisms underlying these isoform-specific effects are unknown. It is possible that myelination requires juxtacrine Nrg1 signaling provided by the membrane-bound isoform, whereas paracrine stimulation by soluble Nrg1 inhibits the process. To investigate this, we asked whether Nrg1 type III provided in a paracrine manner would promote or inhibit myelination. We found that soluble Nrg1 type III enhanced myelination in Schwann cell-neuron cocultures. It improved myelination of Nrg1 type III(+/-) neurons and induced myelination on normally nonmyelinated sympathetic neurons. However, soluble Nrg1 type III failed to induce myelination on Nrg1 type III(-/-) neurons. To our surprise, low concentrations of Nrg1 type II also elicited a similar promyelinating effect. At high doses, however, both type II and III isoforms inhibited myelination and increased c-Jun expression in a manner dependent on Mek/Erk (mitogen-activated protein kinase kinase/extracellular signal-regulated kinase) activation. These results indicate that paracrine Nrg1 signaling provides concentration-dependent bifunctional effects on Schwann cell myelination. Furthermore, our studies suggest that there may be two distinct steps in Schwann cell myelination: an initial phase dependent on juxtacrine Nrg1 signaling and a later phase that can be promoted by paracrine stimulation.


Assuntos
Bainha de Mielina/metabolismo , Neuregulina-1/metabolismo , Células de Schwann/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Gânglios Espinais/enzimologia , Gânglios Espinais/metabolismo , Genes jun , Humanos , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Bainha de Mielina/enzimologia , Neuregulina-1/genética , Neurônios/enzimologia , Neurônios/metabolismo , Isoformas de Proteínas/metabolismo , Ratos , Células de Schwann/enzimologia , Nervo Isquiático/enzimologia , Nervo Isquiático/metabolismo
20.
J Neurosci Res ; 89(3): 310-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21259318

RESUMO

The polarized domains of myelinated axons are specifically organized to maximize the efficiency of saltatory conduction. The paranodal region is directly adjacent to the node of Ranvier and contains specialized septate-like junctions that provide adhesion between axons and glial cells and that constitute a lateral diffusion barrier for nodal components. To complement and extend earlier studies on the peripheral nervous system, electron tomography was used to image paranodal regions from the central nervous system (CNS). Our three-dimensional reconstructions revealed short filamentous linkers running directly from the septate-like junctions to neurofilaments, microfilaments, and organelles within the axon. The intercellular spacing between axons and glia was measured to be 7.4 ± 0.6 nm, over twice the value previously reported in the literature (2.5-3.0 nm). Averaging of individual junctions revealed a bifurcated structure in the intercellular space that is consistent with a dimeric complex of cell adhesion molecules composing the septate-like junction. Taken together, these findings provide new insight into the structural organization of CNS paranodes and suggest that, in addition to providing axo-glial adhesion, cytoskeletal linkage to the septate-like junctions may be required to maintain axonal domains and to regulate organelle transport in myelinated axons.


Assuntos
Axônios/ultraestrutura , Sistema Nervoso Central/citologia , Citoesqueleto/ultraestrutura , Tomografia com Microscopia Eletrônica/métodos , Junções Intercelulares/ultraestrutura , Neuroglia/citologia , Animais , Axônios/metabolismo , Citoesqueleto/metabolismo , Camundongos , Proteínas de Neurofilamentos/metabolismo , Proteínas de Neurofilamentos/ultraestrutura , Neuroglia/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA