Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Exp Eye Res ; 171: 54-61, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29526794

RESUMO

Optic neuropathies such as glaucoma are characterized by the degeneration of retinal ganglion cells (RGCs) and the irreversible loss of vision. In these diseases, focal axon injury triggers a propagating axon degeneration and, eventually, cell death. Previous work by us and others identified dual leucine zipper kinase (DLK) and JUN N-terminal kinase (JNK) as key mediators of somal cell death signaling in RGCs following axonal injury. Moreover, others have shown that activation of the DLK/JNK pathway contributes to distal axonal degeneration in some neuronal subtypes and that this activation is dependent on the adaptor protein, sterile alpha and TIR motif containing 1 (SARM1). Given that SARM1 acts upstream of DLK/JNK signaling in axon degeneration, we tested whether SARM1 plays a similar role in RGC somal apoptosis in response to optic nerve injury. Using the mouse optic nerve crush (ONC) model, our results show that SARM1 is critical for RGC axonal degeneration and that axons rescued by SARM1 deficiency are electrophysiologically active. Genetic deletion of SARM1 did not, however, prevent DLK/JNK pathway activation in RGC somas nor did it prevent or delay RGC cell death. These results highlight the importance of SARM1 in RGC axon degeneration and suggest that somal activation of the DLK/JNK pathway is activated by an as-yet-unidentified SARM1-independent signal.


Assuntos
Proteínas do Domínio Armadillo/fisiologia , Axônios/metabolismo , Proteínas do Citoesqueleto/fisiologia , Modelos Animais de Doenças , Traumatismos do Nervo Óptico/metabolismo , Receptores do Fator de Necrose Tumoral/fisiologia , Degeneração Retiniana/metabolismo , Células Ganglionares da Retina/metabolismo , Animais , Apoptose/fisiologia , Axônios/patologia , Contagem de Células , Sobrevivência Celular , Eletrofisiologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Compressão Nervosa , Traumatismos do Nervo Óptico/patologia , Degeneração Retiniana/patologia , Células Ganglionares da Retina/patologia
2.
J Neurosci Res ; 95(7): 1402-1414, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27614087

RESUMO

Conduction block by nitric oxide (NO) was examined in myelinated and unmyelinated axons from both the central nervous system and peripheral nervous system. In rat vagus nerves, mouse optic nerves at P12-P23, adult and developing mouse sciatic nerves, and mouse spinal cords, myelinated fibers were preferentially blocked reversibly by concentrations of NO similar to those encountered in inflammatory lesions. The possibility that these differences between myelinated and unmyelinated axons are due to the normal developmental substitution of Na+ channel subtype Nav 1.6 for Nav 1.2 at nodes of Ranvier was tested by repeating experiments on mice null for Nav 1.6. Results were unchanged in this mutant. In shiverer optic nerve, which has only scattered regions with nodes of Ranvier, only the fastest component of the compound action potential was reduced. NO was compared with three other methods of blocking conduction: low Na+ , high K+ , and tetrodotoxin (TTX). In each of these three cases, unmyelinated axons lost conduction simultaneously with myelinated fibers. From changes in conduction velocity in myelinated axons as they were blocked, it was ascertained that NO acted most similarly to TTX. It was concluded that NO likely interacts with axonal Na+ channels through an intermediate that is associated with myelin. © 2016 Wiley Periodicals, Inc.


Assuntos
Bloqueio Nervoso Autônomo/métodos , Fibras Nervosas Mielinizadas/fisiologia , Condução Nervosa/fisiologia , Óxido Nítrico/farmacologia , Animais , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Condução Nervosa/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Endogâmicos Lew , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiologia
3.
J Immunol ; 195(6): 2552-9, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26238492

RESUMO

Multiple sclerosis (MS) is believed to be initiated by myelin-reactive CD4(+) Th cells. IL-12-polarized Th1 cells, IL-23-polarized Th17 cells, and Th17 cells that acquire Th1 characteristics were each implicated in autoimmune pathogenesis. It is debated whether Th cells that can drive the development of demyelinating lesions are phenotypically diverse or arise from a single lineage. In the current study, we assessed the requirement of IL-12 or IL-23 stimulation, as well as Th plasticity, for the differentiation of T cells capable of inducing CNS axon damage. We found that stable murine Th1 and Th17 cells independently transfer experimental autoimmune encephalomyelitis (widely used as an animal model of MS) in the absence of IL-23 and IL-12, respectively. Plastic Th17 cells are particularly potent mediators of demyelination and axonopathy. In parallel studies, we identified MS patients who consistently mount either IFN-γ- or IL-17-skewed responses to myelin basic protein over the course of a year. Brain magnetic resonance imaging revealed that patients with mixed IFN-γ and IL-17 responses have relatively high T1 lesion burden, a measure of permanent axon damage. Our data challenge the dogma that IL-23 and Th17 plasticity are universally required for the development of experimental autoimmune encephalomyelitis. This study definitively demonstrates that autoimmune demyelinating disease can be driven by distinct Th-polarizing factors and effector subsets, underscoring the importance of a customized approach to the pharmaceutical management of MS.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/imunologia , Células Th1/imunologia , Células Th17/imunologia , Transferência Adotiva , Animais , Autoimunidade/imunologia , Encéfalo/diagnóstico por imagem , Diferenciação Celular/imunologia , Doenças Desmielinizantes/imunologia , Humanos , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-12/imunologia , Interleucina-17/imunologia , Interleucina-23/imunologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Básica da Mielina/imunologia , Nervo Óptico/imunologia , Nervo Óptico/patologia , Radiografia , Células Th1/citologia , Células Th1/transplante , Células Th17/citologia , Células Th17/transplante
4.
Neurobiol Dis ; 89: 36-45, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26807988

RESUMO

Mutations of the neuronal sodium channel gene SCN8A are associated with lethal movement disorders in the mouse and with human epileptic encephalopathy. We describe a spontaneous mouse mutation, Scn8a(9J), that is associated with a chronic movement disorder with early onset tremor and adult onset dystonia. Scn8a(9J) homozygotes have a shortened lifespan, with only 50% of mutants surviving beyond 6 months of age. The 3 bp in-frame deletion removes 1 of the 3 adjacent isoleucine residues in transmembrane segment DIVS6 of Nav1.6 (p.Ile1750del). The altered helical orientation of the transmembrane segment displaces pore-lining amino acids with important roles in channel activation and inactivation. The predicted impact on channel activity was confirmed by analysis of cerebellar Purkinje neurons from mutant mice, which lack spontaneous and induced repetitive firing. In a heterologous expression system, the activity of the mutant channel was below the threshold for detection. Observations of decreased nerve conduction velocity and impaired behavior in an open field are also consistent with reduced activity of Nav1.6. The Nav1.6Δ1750 protein is only partially glycosylated. The abundance of mutant Nav1.6 is reduced at nodes of Ranvier and is not detectable at the axon initial segment. Despite a severe reduction in channel activity, the lifespan and motor function of Scn8a(9J/9J) mice are significantly better than null mutants lacking channel protein. The clinical phenotype of this severe hypomorphic mutant expands the spectrum of Scn8a disease to include a recessively inherited, chronic and progressive movement disorder.


Assuntos
Aminoácidos/genética , Transtornos dos Movimentos/genética , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Deleção de Sequência , Potenciais de Ação , Animais , Segmento Inicial do Axônio/metabolismo , Comportamento Animal , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiologia , Distonia/complicações , Distonia/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transtornos dos Movimentos/complicações , Transtornos dos Movimentos/veterinária , Força Muscular , Canal de Sódio Disparado por Voltagem NAV1.6/fisiologia , Condução Nervosa , Junção Neuromuscular/patologia , Células de Purkinje/metabolismo , Células de Purkinje/fisiologia , Nós Neurofibrosos/metabolismo , Análise de Sobrevida , Tremor/complicações , Tremor/genética
5.
Neurobiol Dis ; 69: 108-16, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24878510

RESUMO

Injury to retinal ganglion cell (RGC) axons triggers rapid activation of Jun N-terminal kinase (JNK) signaling, a major prodeath pathway in injured RGCs. Of the multiple kinases that can activate JNK, dual leucine kinase (Dlk) is known to regulate both apoptosis and Wallerian degeneration triggered by axonal insult. Here we tested the importance of Dlk in regulating somal and axonal degeneration of RGCs following axonal injury. Removal of DLK from the developing optic cup did not grossly affect developmental RGC death or inner plexiform layer organization. In the adult, Dlk deficiency significantly delayed axonal-injury induced RGC death. The activation of JUN was also attenuated in Dlk deficient retinas. Dlk deficiency attenuated the activation of the somal pool of JNK but did not prevent activation of the axonal pool of JNK after axonal injury, indicating that JNK activation in different cellular compartments of an RGC following axonal injury is regulated by distinct upstream kinases. In contrast to its robust influence on somal degeneration, Dlk deficiency did not alter RGC axonal degeneration after axonal injury as assessed using physiological readouts of optic nerve function.


Assuntos
Axônios/enzimologia , MAP Quinase Quinase Quinases/deficiência , Traumatismos do Nervo Óptico/enzimologia , Células Ganglionares da Retina/enzimologia , Degeneração Walleriana/enzimologia , Animais , Axônios/patologia , Morte Celular/fisiologia , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinases/genética , Camundongos Transgênicos , Traumatismos do Nervo Óptico/patologia , Fosforilação/fisiologia , Retina/enzimologia , Retina/crescimento & desenvolvimento , Retina/patologia , Células Ganglionares da Retina/patologia , Transdução de Sinais , Técnicas de Cultura de Tecidos , Degeneração Walleriana/patologia
6.
J Nutr ; 144(7): 1058-66, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24744313

RESUMO

Gestational iron deficiency (ID) has been associated with a wide variety of central nervous system (CNS) impairments in developing offspring. However, a focus on singular regions has impeded an understanding of the CNS-wide effects of this micronutrient deficiency. Because the developing brain requires iron during specific phases of growth in a region-specific manner, we hypothesized that maternal iron deprivation would lead to region-specific impairments in the CNS of offspring. Female rats were fed an iron control (Fe+) or iron-deficient (Fe-) diet containing 240 or 6 µg/g iron during gestation and lactation. The corpus callosum (CC), hippocampus, and cortex of the offspring were analyzed at postnatal day 21 (P21) and/or P40 using structural and functional measures. In the CC at P40, ID was associated with reduced peak amplitudes of compound action potentials specific to myelinated axons, in which diameters were reduced by ∼20% compared with Fe+ controls. In the hippocampus, ID was associated with a 25% reduction in basal dendritic length of pyramidal neurons at P21, whereas branching complexity was unaffected. We also identified a shift toward increased proximal branching of apical dendrites in ID without an effect on overall length compared with Fe+ controls. ID also affected cortical neurons, but unlike the hippocampus, both apical and basal dendrites displayed a uniform decrease in branching complexity, with no significant effect on overall length. These deficits culminated in significantly poorer performance of P40 Fe- offspring in the novel object recognition task. Collectively, these results demonstrate that non-anemic gestational ID has a significant and region-specific impact on neuronal development and may provide a framework for understanding and recognizing the presentation of clinical symptoms of ID.


Assuntos
Dano Encefálico Crônico/etiologia , Córtex Cerebral/diagnóstico por imagem , Corpo Caloso/diagnóstico por imagem , Deficiências de Ferro , Lactação , Fenômenos Fisiológicos da Nutrição Materna , Neurônios/diagnóstico por imagem , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Dano Encefálico Crônico/congênito , Dano Encefálico Crônico/metabolismo , Dano Encefálico Crônico/patologia , Córtex Cerebral/metabolismo , Corpo Caloso/metabolismo , Dendritos/metabolismo , Dendritos/ultraestrutura , Feminino , Hipocampo/diagnóstico por imagem , Hipocampo/metabolismo , Masculino , Fibras Nervosas Mielinizadas/diagnóstico por imagem , Fibras Nervosas Mielinizadas/metabolismo , Neurogênese , Neurônios/metabolismo , Gravidez , Células Piramidais/diagnóstico por imagem , Células Piramidais/metabolismo , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Reprodutibilidade dos Testes , Ultrassonografia
7.
bioRxiv ; 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38766050

RESUMO

Precise regulation of protein phosphorylation is critical for many cellular processes, and dysfunction in this process has been linked to various neurological disorders and diseases. Protein phosphatase 1 (PP1) is a ubiquitously expressed serine/threonine phosphatase with three major isoforms, (α, ß, γ) and hundreds of known substrates. Previously, we reported that PP1α and PP1γ are essential for the known role of PP1 in synaptic physiology and learning/memory, while PP1ß displayed a surprising opposing function. De novo mutations in PP1ß cause neurodevelopmental disorders in humans, but the mechanisms involved are currently unknown. A Cre-Lox system was used to delete PP1ß specifically in neurons in order to study its effects on developing mice. These animals fail to survive to 3 postnatal weeks, and exhibit deficits in cortical myelination and glutamate release. There was defective compound action potential (CAP) propagation in the optic nerve of the null mice, which was traced to a deficit in the formation of nodes of Ranvier. Finally, it was found that phosphorylation of the PP1ß-specific substrate, myosin light chain 2 (MLC2), is significantly enhanced in PP1ß null optic nerves. Several novel important in vivo roles of PP1ß in neurons were discovered, and these data will aid future investigations in delineating the mechanisms by which de novo mutations in PP1ß lead to intellectual and developmental delays in patients.

8.
bioRxiv ; 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38496662

RESUMO

Upon peripheral nervous system (PNS) injury, severed axons undergo rapid SARM1-dependent Wallerian degeneration (WD). In mammals, the role of SARM1 in PNS regeneration, however, is unknown. Here we demonstrate that Sarm1 is not required for axotomy induced activation of neuron-intrinsic growth programs and axonal growth into a nerve crush site. However, in the distal nerve, Sarm1 is necessary for the timely induction of the Schwann cell (SC) repair response, nerve inflammation, myelin clearance, and regeneration of sensory and motor axons. In Sarm1-/- mice, regenerated fibers exhibit reduced axon caliber, defective nerve conduction, and recovery of motor function is delayed. The growth hostile environment of Sarm1-/- distal nerve tissue was demonstrated by grafting of Sarm1-/- nerve into WT recipients. SC lineage tracing in injured WT and Sarm1-/- mice revealed morphological differences. In the Sarm1-/- distal nerve, the appearance of p75NTR+, c-Jun+ SCs is significantly delayed. Ex vivo, p75NTR and c-Jun upregulation in Sarm1-/- nerves can be rescued by pharmacological inhibition of ErbB kinase. Together, our studies show that Sarm1 is not necessary for the activation of neuron intrinsic growth programs but in the distal nerve is required for the orchestration of cellular programs that underlie rapid axon extension.

9.
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
10.
J Neurosci ; 31(48): 17736-51, 2011 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-22131434

RESUMO

The plt (pale tremor) mouse carries a null mutation in the Fig4(Sac3) gene that results in tremor, hypopigmentation, spongiform degeneration of the brain, and juvenile lethality. FIG4 is a ubiquitously expressed phosphatidylinositol 3,5-bisphosphate phosphatase that regulates intracellular vesicle trafficking along the endosomal-lysosomal pathway. In humans, the missense mutation FIG4(I41T) combined with a FIG4 null allele causes Charcot-Marie-Tooth 4J disease, a severe form of peripheral neuropathy. Here we show that Fig4 null mice exhibit a dramatic reduction of myelin in the brain and spinal cord. In the optic nerve, smaller-caliber axons lack myelin sheaths entirely, whereas many large- and intermediate-caliber axons are myelinated but show structural defects at nodes of Ranvier, leading to delayed propagation of action potentials. In the Fig4 null brain and optic nerve, oligodendrocyte (OL) progenitor cells are present at normal abundance and distribution, but the number of myelinating OLs is greatly compromised. The total number of axons in the Fig4 null optic nerve is not reduced. Developmental studies reveal incomplete myelination rather than elevated cell death in the OL linage. Strikingly, there is rescue of CNS myelination and tremor in transgenic mice with neuron-specific expression of Fig4, demonstrating a non-cell-autonomous function of Fig4 in OL maturation and myelin development. In transgenic mice with global overexpression of the human pathogenic FIG4 variant I41T, there is rescue of the myelination defect, suggesting that the CNS of CMT4J patients may be protected from myelin deficiency by expression of the FIG4(I41T) mutant protein.


Assuntos
Axônios/patologia , Doença de Charcot-Marie-Tooth/genética , Flavoproteínas/genética , Bainha de Mielina/genética , Fibras Nervosas Mielinizadas/patologia , Neurônios/metabolismo , Monoéster Fosfórico Hidrolases/genética , Animais , Axônios/metabolismo , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Flavoproteínas/metabolismo , Camundongos , Camundongos Transgênicos , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Fibras Nervosas Mielinizadas/metabolismo , Neurônios/patologia , Fosfatidilinositóis/metabolismo , Fosfatases de Fosfoinositídeos , Monoéster Fosfórico Hidrolases/metabolismo
11.
J Cell Biol ; 178(5): 875-86, 2007 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-17709431

RESUMO

Action potential initiation and propagation requires clustered Na(+) (voltage-gated Na(+) [Nav]) channels at axon initial segments (AIS) and nodes of Ranvier. In addition to ion channels, these domains are characterized by cell adhesion molecules (CAMs; neurofascin-186 [NF-186] and neuron glia-related CAM [NrCAM]), cytoskeletal proteins (ankyrinG and betaIV spectrin), and the extracellular chondroitin-sulfate proteoglycan brevican. Schwann cells initiate peripheral nervous system node formation by clustering NF-186, which then recruits ankyrinG and Nav channels. However, AIS assembly of this protein complex does not require glial contact. To determine the AIS assembly mechanism, we silenced expression of AIS proteins by RNA interference. AnkyrinG knockdown prevented AIS localization of all other AIS proteins. Loss of NF-186, NrCAM, Nav channels, or betaIV spectrin did not affect other neuronal AIS proteins. However, loss of NF-186 blocked assembly of the brevican-based AIS extracellular matrix, and NF-186 overexpression caused somatodendritic brevican clustering. Thus, NF-186 assembles and links the specialized brevican-containing AIS extracellular matrix to the intracellular cytoskeleton.


Assuntos
Axônios , Moléculas de Adesão Celular/metabolismo , Matriz Extracelular/metabolismo , Fatores de Crescimento Neural/metabolismo , Animais , Anquirinas/genética , Anquirinas/metabolismo , Axônios/metabolismo , Axônios/ultraestrutura , Brevicam , Moléculas de Adesão Celular/genética , Células Cultivadas , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Citoesqueleto/metabolismo , Matriz Extracelular/química , Hipocampo/citologia , Lectinas Tipo C/metabolismo , Fatores de Crescimento Neural/genética , Proteínas do Tecido Nervoso/metabolismo , RNA/genética , RNA/metabolismo , Interferência de RNA , Nós Neurofibrosos/metabolismo , Nós Neurofibrosos/ultraestrutura , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Canais de Sódio/genética , Canais de Sódio/metabolismo , Espectrina/genética , Espectrina/metabolismo
12.
Mol Neurobiol ; 59(12): 7486-7494, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36198882

RESUMO

Nuclear inhibitor of protein phosphatase 1 (NIPP1) is a known regulator of gene expression and plays roles in many physiological or pathological processes such as stem cell proliferation and skin inflammation. While NIPP1 has many regulatory roles in proliferating cells, its function in the central nervous system (CNS) has not been directly investigated. In the present study, we examined NIPP1 CNS function using a conditional knockout (cKO) mouse model in which the Nipp1 gene is excised from neural precursor cells. These mice exhibited severe developmental impairments that led to premature lethality. To delineate the neurological changes occurring in these animals, we first assessed microtubule-associated protein tau, a known target of NIPP1 activity. We found that phosphorylation of tau is significantly enhanced in NIPP1 cKO mice. Consistent with this, we found altered AKT and PP1 activity in NIPP1 cKO mice, suggesting that increased tau phosphorylation likely results from a shift in kinase/phosphatase activity. Secondly, we observed tremors in the NIPP1 cKO mice which prompted us to explore the integrity of the myelin sheath, an integral structure for CNS function. We demonstrated that in NIPP1 cKO mice, there is a significant decrease in MBP protein expression in the cortex, along with deficits in both the conduction of compound action potentials (CAP) and the percentage of myelinated axons in the optic nerve. Our study suggests that NIPP1 in neural precursor cells regulates phosphorylation of tau and CNS myelination and may represent a novel therapeutic target for neurodegenerative diseases.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Células-Tronco Neurais , Camundongos , Animais , Proteína Fosfatase 1/metabolismo , Fosforilação , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células-Tronco Neurais/metabolismo , Sistema Nervoso Central/metabolismo , Bainha de Mielina/metabolismo
13.
J Neurosci ; 30(37): 12432-45, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20844138

RESUMO

In the adult mammalian CNS, the growth inhibitors oligodendrocyte-myelin glycoprotein (OMgp) and the reticulon RTN4 (Nogo) are broadly expressed in oligodendrocytes and neurons. Nogo and OMgp complex with the neuronal cell surface receptors Nogo receptor-1 (NgR1) and paired Ig-like receptor-B (PirB) to regulate neuronal morphology. In the healthy CNS, NgR1 regulates dendritic spine shape and attenuates activity-driven synaptic plasticity at Schaffer collateral-CA1 synapses. Here, we examine whether Nogo and OMgp influence functional synaptic plasticity, the efficacy by which synaptic transmission occurs. In acute hippocampal slices of adult mice, Nogo-66 and OMgp suppress NMDA receptor-dependent long-term potentiation (LTP) when locally applied to Schaffer collateral-CA1 synapses. Neither Nogo-66 nor OMgp influences basal synaptic transmission or paired-pulse facilitation, a form of short-term synaptic plasticity. PirB(-/-) and NgR1(-/-) single mutants and NgR1(-/-);PirB(-/-) double mutants show normal LTP, indistinguishable from wild-type controls. In juvenile mice, LTD in NgR1(-/-), but not PirB(-/-), slices is absent. Mechanistic studies revealed that Nogo-66 and OMgp suppress LTP in an NgR1-dependent manner. OMgp inhibits LTP in part through PirB but independently of p75. This suggests that NgR1 and PirB participate in ligand-dependent inhibition of synaptic plasticity. Loss of NgR1 leads to increased phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), signaling intermediates known to regulate neuronal growth and synaptic function. In primary cortical neurons, BDNF elicited phosphorylation of AKT and p70S6 kinase is attenuated in the presence of myelin inhibitors. Collectively, we provide evidence that mechanisms of neuronal growth inhibition and inhibition of synaptic strength are related. Thus, myelin inhibitors and their receptors may coordinate structural and functional neuronal plasticity in CNS health and disease.


Assuntos
Regulação para Baixo/fisiologia , Proteínas da Mielina/fisiologia , Glicoproteína Associada a Mielina/fisiologia , Inibição Neural/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Linhagem Celular , Regulação para Baixo/genética , Proteínas Ligadas por GPI , Humanos , Potenciação de Longa Duração/genética , Potenciação de Longa Duração/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Glicoproteína Associada a Mielina/genética , Glicoproteína Associada a Mielina/farmacologia , Glicoproteína Mielina-Oligodendrócito , Inibição Neural/genética , Plasticidade Neuronal/genética , Proteínas Nogo , Receptor Nogo 1 , Ratos , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/fisiologia , Receptores Imunológicos/deficiência , Receptores Imunológicos/metabolismo , Receptores Imunológicos/fisiologia
14.
Neuron ; 47(5): 681-94, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16129398

RESUMO

The signals that determine whether axons are ensheathed or myelinated by Schwann cells have long been elusive. We now report that threshold levels of neuregulin-1 (NRG1) type III on axons determine their ensheathment fate. Ensheathed axons express low levels whereas myelinated fibers express high levels of NRG1 type III. Sensory neurons from NRG1 type III deficient mice are poorly ensheathed and fail to myelinate; lentiviral-mediated expression of NRG1 type III rescues these defects. Expression also converts the normally unmyelinated axons of sympathetic neurons to myelination. Nerve fibers of mice haploinsufficient for NRG1 type III are disproportionately unmyelinated, aberrantly ensheathed, and hypomyelinated, with reduced conduction velocities. Type III is the sole NRG1 isoform retained at the axon surface and activates PI 3-kinase, which is required for Schwann cell myelination. These results indicate that levels of NRG1 type III, independent of axon diameter, provide a key instructive signal that determines the ensheathment fate of axons.


Assuntos
Axônios/fisiologia , Bainha de Mielina/fisiologia , Neuregulina-1/fisiologia , Potenciais de Ação/fisiologia , Animais , Contagem de Células , Tamanho Celular , Células Cultivadas , Detergentes/química , Eletrofisiologia , Feminino , Imunofluorescência , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Genótipo , Lentivirus/crescimento & desenvolvimento , Metaloproteases , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Neuritos/fisiologia , Sistema Nervoso Periférico/citologia , Sistema Nervoso Periférico/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Gravidez , Ratos , Células de Schwann/fisiologia , Transdução de Sinais
15.
J Neurosci ; 28(11): 2753-65, 2008 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-18337405

RESUMO

In the mature nervous system, changes in synaptic strength correlate with changes in neuronal structure. Members of the Nogo-66 receptor family have been implicated in regulating neuronal morphology. Nogo-66 receptor 1 (NgR1) supports binding of the myelin inhibitors Nogo-A, MAG (myelin-associated glycoprotein), and OMgp (oligodendrocyte myelin glycoprotein), and is important for growth cone collapse in response to acutely presented inhibitors in vitro. After injury to the corticospinal tract, NgR1 limits axon collateral sprouting but is not important for blocking long-distance regenerative growth in vivo. Here, we report on a novel interaction between NgR1 and select members of the fibroblast growth factor (FGF) family. FGF1 and FGF2 bind directly and with high affinity to NgR1 but not to NgR2 or NgR3. In primary cortical neurons, ectopic NgR1 inhibits FGF2-elicited axonal branching. Loss of NgR1 results in altered spine morphologies along apical dendrites of hippocampal CA1 neurons in vivo. Analysis of synaptosomal fractions revealed that NgR1 is enriched synaptically in the hippocampus. Physiological studies at Schaffer collateral-CA1 synapses uncovered a synaptic function for NgR1. Loss of NgR1 leads to FGF2-dependent enhancement of long-term potentiation (LTP) without altering basal synaptic transmission or short-term plasticity. NgR1 and FGF receptor 1 (FGFR1) are colocalized to synapses, and mechanistic studies revealed that FGFR kinase activity is necessary for FGF2-elicited enhancement of hippocampal LTP in NgR1 mutants. In addition, loss of NgR1 attenuates long-term depression of synaptic transmission at Schaffer collateral-CA1 synapses. Together, our findings establish that physiological NgR1 signaling regulates activity-dependent synaptic strength and uncover neuronal NgR1 as a regulator of synaptic plasticity.


Assuntos
Espinhas Dendríticas/fisiologia , Receptores de Superfície Celular/fisiologia , Sinapses/fisiologia , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Espinhas Dendríticas/ultraestrutura , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteínas Ligadas por GPI , Humanos , Camundongos , Camundongos Mutantes , Receptor Nogo 2 , Ligação Proteica/fisiologia , Ratos , Sinapses/ultraestrutura
16.
J Cell Biol ; 162(6): 1149-60, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12963709

RESUMO

In myelinated axons, K+ channels are concealed under the myelin sheath in the juxtaparanodal region, where they are associated with Caspr2, a member of the neurexin superfamily. Deletion of Caspr2 in mice by gene targeting revealed that it is required to maintain K+ channels at this location. Furthermore, we show that the localization of Caspr2 and clustering of K+ channels at the juxtaparanodal region depends on the presence of TAG-1, an immunoglobulin-like cell adhesion molecule that binds Caspr2. These results demonstrate that Caspr2 and TAG-1 form a scaffold that is necessary to maintain K+ channels at the juxtaparanodal region, suggesting that axon-glia interactions mediated by these proteins allow myelinating glial cells to organize ion channels in the underlying axonal membrane.


Assuntos
Axônios/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas de Membrana , Fibras Nervosas Mielinizadas/metabolismo , Proteínas do Tecido Nervoso/deficiência , Canais de Potássio/metabolismo , Nós Neurofibrosos/metabolismo , Animais , Axônios/ultraestrutura , Comunicação Celular/genética , Contactina 2 , Marcação de Genes , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Mutação/genética , Fibras Nervosas Mielinizadas/ultraestrutura , Proteínas do Tecido Nervoso/genética , Condução Nervosa/genética , Neuroglia/metabolismo , Neuroglia/ultraestrutura , Canais de Potássio/genética , Nós Neurofibrosos/ultraestrutura , Superfamília Shaker de Canais de Potássio
17.
Elife ; 62017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29251594

RESUMO

Low-density lipoprotein receptor-related protein-1 (LRP1) is a large endocytic and signaling molecule broadly expressed by neurons and glia. In adult mice, global inducible (Lrp1flox/flox;CAG-CreER) or oligodendrocyte (OL)-lineage specific ablation (Lrp1flox/flox;Pdgfra-CreER) of Lrp1 attenuates repair of damaged white matter. In oligodendrocyte progenitor cells (OPCs), Lrp1 is required for cholesterol homeostasis and differentiation into mature OLs. Lrp1-deficient OPC/OLs show a strong increase in the sterol-regulatory element-binding protein-2 yet are unable to maintain normal cholesterol levels, suggesting more global metabolic deficits. Mechanistic studies revealed a decrease in peroxisomal biogenesis factor-2 and fewer peroxisomes in OL processes. Treatment of Lrp1-/- OPCs with cholesterol or activation of peroxisome proliferator-activated receptor-γ with pioglitazone alone is not sufficient to promote differentiation; however, when combined, cholesterol and pioglitazone enhance OPC differentiation into mature OLs. Collectively, our studies reveal a novel role for Lrp1 in peroxisome biogenesis, lipid homeostasis, and OPC differentiation during white matter development and repair.


Assuntos
Colesterol/metabolismo , Homeostase , Bainha de Mielina/metabolismo , Células Precursoras de Oligodendrócitos/fisiologia , Biogênese de Organelas , Peroxissomos/metabolismo , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos
19.
Mol Neurodegener ; 12(1): 71, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28969695

RESUMO

BACKGROUND: Optic nerve injury is an important pathological component in neurodegenerative diseases such as traumatic optic neuropathies and glaucoma. The molecular signaling pathway(s) critical for retinal ganglion cell (RGC) death after axonal insult, however, is/are not fully defined. RGC death after axonal injury is known to occur by BAX-dependent apoptosis. Two transcription factors JUN (the canonical target of JNK) and DDIT3 (CHOP; a key mediator of the endoplasmic reticulum stress response) are known to be important apoptotic signaling molecules after axonal injury, including in RGCs. However, neither Jun nor Ddit3 deficiency provide complete protection to RGCs after injury. Since Jun and Ddit3 are important apoptotic signaling molecules, we sought to determine if their combined deficiency might provide additive protection to RGCs after axonal injury. METHODS: To determine if DDIT3 regulated the expression of JUN after an axonal insult, mice deficient for Ddit3 were examined after optic nerve crush (ONC). In order to critically test the importance of these genes in RGC death after axonal injury, RGC survival was assessed at multiple time-points after ONC (14, 35, 60, and 120 days after injury) in Jun, Ddit3, and combined Jun/Ddit3 deficient mice. Finally, to directly assess the role of JUN and DDIT3 in axonal degeneration, compound actions potentials were recorded from Jun, Ddit3, and Jun/Ddit3 deficient mice after ONC. RESULTS: Single and combined deficiency of Jun and Ddit3 did not appear to alter gross retinal morphology. Ddit3 deficiency did not alter expression of JUN after axonal injury. Deletion of both Jun and Ddit3 provided significantly greater long-term protection to RGCs as compared to Jun or Ddit3 deficiency alone. Finally, despite the profound protection to RGC somas provided by the deficiency of Jun plus Ddit3, their combined loss did not lessen axonal degeneration. CONCLUSIONS: These results suggest JUN and DDIT3 are independently regulated pro-death signaling molecules in RGCs and together account for the vast majority of apoptotic signaling in RGCs after axonal injury. Thus, JUN and DDIT3 may represent key molecular hubs that integrate upstream signaling events triggered by axonal injury with downstream transcriptional events that ultimately culminate in RGC apoptosis.


Assuntos
Genes jun/fisiologia , Degeneração Neural/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Células Ganglionares da Retina/patologia , Fator de Transcrição CHOP/metabolismo , Animais , Apoptose/fisiologia , Axônios/metabolismo , Axônios/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Compressão Nervosa , Degeneração Neural/patologia , Células Ganglionares da Retina/metabolismo , Transdução de Sinais/fisiologia
20.
Int J Radiat Oncol Biol Phys ; 96(2): 449-457, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27478169

RESUMO

PURPOSE: To determine the late effects of fractionated versus single-dose cranial radiation on murine white matter. METHODS AND MATERIALS: Mice were exposed to 0 Gy, 6 × 6 Gy, or 1 × 20 Gy cranial irradiation at 10 to 12 weeks of age. Endpoints were assessed through 18 months from exposure using immunohistochemistry, electron microscopy, and electrophysiology. RESULTS: Weight gain was temporarily reduced after irradiation; greater loss was seen after single versus fractionated doses. Oligodendrocyte progenitor cells were reduced early and late after both single and fractionated irradiation. Both protocols also increased myelin g-ratio, reduced the number of nodes of Ranvier, and promoted a shift in the proportion of small, unmyelinated versus large, myelinated axon fibers. CONCLUSIONS: Fractionation does not adequately spare normal white matter from late radiation side effects.


Assuntos
Linhagem da Célula/efeitos da radiação , Irradiação Craniana/efeitos adversos , Fracionamento da Dose de Radiação , Oligodendroglia/efeitos da radiação , Aumento de Peso/efeitos da radiação , Substância Branca/efeitos da radiação , Animais , Células Cultivadas , Relação Dose-Resposta à Radiação , Camundongos , Oligodendroglia/patologia , Tratamentos com Preservação do Órgão/métodos , Órgãos em Risco/efeitos da radiação , Doses de Radiação , Proteção Radiológica/métodos , Substância Branca/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA