Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 35(10): 886-891, 2019 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-31814564

RESUMO

Objective To establish and optimize the prokaryotic expression method for the recombinant mouse myelin proteolipid protein (PLP, 139-208 aa) which is a critical immunogenic polypeptide of PLP. Methods The sequence coding for PLP139-208 polypeptide was cloned into pET-32a(+) vector. Afterwards, the expression vector prepared in this research was transformed into E. coli BL21, and the recombinant PLP polypeptide was induced to express by isopropyl-ß-D-thiogalactoside (IPTG). Two key prokaryotic expression conditions, IPTG's induction length and temperature, were analyzed for further optimization. The recombinant PLP polypeptide was induced to express by the expression method under the optimal expression conditions, and then was purified by Ni-NTA agarose and amylose resin. Finally, the gain of PLP139-208 polypeptide was verified by Western blot analysis. Results The results in the combinatorial optimization revealed that the expression of PLP139-208 was obtained at a satisfactory level when it was incubated at 23DegreesCelsius for 20 hours with the IPTG concentration of 0.5 mmol/L. Conclusion The optimized prokaryotic expression method for the recombinant mouse PLP139-208 was successfully established and effectively performed. This will shed light on the further researches on the improved preparation for experimental autoimmune encephalitis (EAE, an animal model of multiple sclerosis) and the underlying mechanism underlying PLP-induced autoimmune demyelination.


Assuntos
Proteína Proteolipídica de Mielina/biossíntese , Animais , Escherichia coli , Isopropiltiogalactosídeo , Camundongos , Peptídeos , Proteínas Recombinantes/biossíntese
2.
Acta Neurobiol Exp (Wars) ; 79(4): 367-373, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31885393

RESUMO

The national incidence of neonatal abstinence syndrome has dramatically increased over the last decade due to an increase in antenatal opioid exposure. Recent human and animal studies suggest that antenatal opioid exposure impacts the developing brain. The purpose of this study is to evaluate the effects of perinatal methadone exposure on myelination in multiple regions in the developing rat brain. Pregnant Sprague-Dawley rats were randomly assigned into three experimental groups and subsequently exposed to drinking water alone or drinking water containing methadone from 7 days post coitum through day 7 or through day 19 after delivery. Two male neonatal rats were randomly selected from each litter and terminated at day 19. The cerebral cortex, hippocampus, cerebellum, and brainstem were dissected and analyzed for three myelin specific proteins - CNP, PLP, and MBP - by Western blot analysis. All pups with exposure to methadone demonstrated decreased expression of CNP, PLP, and MBP in the cerebral cortex and hippocampus. In the cerebellum, PLP expression was down­regulated without apparent alteration of CNP and MBP expression. PLP and MBP expression, but not CNP expression, were significantly inhibited in the brainstem. Compared to the pups with postnatal methadone exposure via maternal milk through day 7, partial recovery of CNP and PLP expression only occurred in the cerebral cortices of the pups exposed through day 19. The findings show that antenatal opioid exposure in rat pups is associated with regionally­specific alterations in brain myelination that diversely affects myelin proteins.


Assuntos
2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/biossíntese , Encéfalo/efeitos dos fármacos , Metadona/toxicidade , Proteína Básica da Mielina/biossíntese , Proteína Proteolipídica de Mielina/biossíntese , Síndrome de Abstinência Neonatal/metabolismo , Efeitos Tardios da Exposição Pré-Natal , 2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/genética , Animais , Encéfalo/embriologia , Feminino , Masculino , Proteína Básica da Mielina/genética , Proteína Proteolipídica de Mielina/genética , Bainha de Mielina/fisiologia , Síndrome de Abstinência Neonatal/etiologia , Oligodendroglia/metabolismo , Especificidade de Órgãos , Gravidez , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
3.
Stem Cells ; 34(4): 984-96, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26676415

RESUMO

Pelizaeus-Merzbacher disease (PMD) results from an X-linked misexpression of proteolipid protein 1 (PLP1). This leukodystrophy causes severe hypomyelination with progressive inflammation, leading to neurological dysfunctions and shortened life expectancy. While no cure exists for PMD, experimental cell-based therapy in the dysmyelinated shiverer model suggested that human oligodendrocyte progenitor cells (hOPCs) or human neural precursor cells (hNPCs) are promising candidates to treat myelinopathies. However, the fate and restorative advantages of human NPCs/OPCs in a relevant model of PMD has not yet been addressed. Using a model of Plp1 overexpression, resulting in demyelination with progressive inflammation, we compared side-by-side the therapeutic benefits of intracerebrally grafted hNPCs and hOPCs. Our findings reveal equal integration of the donor cells within presumptive white matter tracks. While the onset of exogenous remyelination was earlier in hOPCs-grafted mice than in hNPC-grafted mice, extended lifespan occurred only in hNPCs-grafted animals. This improved survival was correlated with reduced neuroinflammation (microglial and astrocytosis loads) and microglia polarization toward M2-like phenotype followed by remyelination. Thus modulation of neuroinflammation combined with myelin restoration is crucial to prevent PMD pathology progression and ensure successful rescue of PMD mice. These findings should help to design novel therapeutic strategies combining immunomodulation and stem/progenitor cell-based therapy for disorders associating hypomyelination with inflammation as observed in PMD.


Assuntos
Imunidade Inata , Inflamação/terapia , Células-Tronco Neurais/transplante , Oligodendroglia/transplante , Doença de Pelizaeus-Merzbacher/terapia , Animais , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imunomodulação , Inflamação/imunologia , Inflamação/patologia , Camundongos , Microglia/imunologia , Microglia/patologia , Proteína Proteolipídica de Mielina/biossíntese , Bainha de Mielina/metabolismo , Células-Tronco Neurais/imunologia , Oligodendroglia/imunologia , Doença de Pelizaeus-Merzbacher/imunologia , Doença de Pelizaeus-Merzbacher/patologia , Regeneração
4.
FASEB J ; 27(5): 1868-73, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23335052

RESUMO

ß-Site amyloid precursor protein convertase enzyme 1 (BACE1), a type I transmembrane aspartyl protease required to cleave amyloid precursor protein for releasing a toxic amyloid peptide, also cleaves type I and type III neuregulin-1 (Nrg-1). BACE1 deficiency in mice causes hypomyelination during development and impairs remyelination if injured. In BACE1-null mice, the abolished cleavage of neuregulin-1 by BACE1 is speculated to cause reduced myelin sheath thickness in both the central nervous system and peripheral nervous system because reduced cleavage of Nrg-1 correlates with reduced Akt phosphorylation, a downstream signaling molecule of the Nrg-1/ErbB pathway. Here we tested specifically whether increasing Akt activity alone in oligodendrocytes would be sufficient to reverse the hypomyelination phenotype in BACE1-null mice. BACE1-null mice were bred with transgenic mice expressing constitutively active Akt (Akt-DD; mutations with D(308)T and D(473)S) in oligodendrocytes. Relative to littermate BACE1-null controls, BACE1(-/-)/Akt-DD mice exhibited enhanced expression of myelin basic protein and promoter of proteolipid protein. The elevated expression of myelin proteins correlated with a thicker myelin sheath in optic nerves; comparison of quantified g ratios with statistic significance was used to confirm this reversion. However, it appeared that myelin sheath thickness in the sciatic nerves was not increased in BACE1(-/-)/Akt-DD mice, as the g ratio was not significantly different from the control. Hence, increased Akt activity in BACE1-null myelinating cells only compensates for the loss of BACE1 activity in the central nervous system, which is consistent with the observation that overexpression of Akt-DD in Schwann cells did not induce hypermyelination. Our results suggest that signaling activity other than Akt may also contribute to proper myelination in peripheral nerves.


Assuntos
Secretases da Proteína Precursora do Amiloide/deficiência , Ácido Aspártico Endopeptidases/deficiência , Proteína Básica da Mielina/biossíntese , Proteína Proteolipídica de Mielina/biossíntese , Bainha de Mielina/fisiologia , Proteínas Proto-Oncogênicas c-akt/biossíntese , Animais , Feminino , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Bainha de Mielina/patologia , Oligodendroglia/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-akt/genética
5.
J Neurosci Res ; 90(9): 1701-12, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22504928

RESUMO

Overexpression of the major myelin proteolipid protein (PLP) is detrimental to brain development and function and is the most common cause of Pelizaeus-Merzbacher disease. microRNA (miRNA), small, noncoding RNAs, have been shown to play critical roles in oligodendrocyte lineage. In this study, we sought to investigate whether miRNAs control PLP abundance. To identify candidate miRNAs involved in this regulation, we have examined differentiation-induced changes in the expression of miRNAs in the oligodendroglial cell line Oli-neu and in enhanced green fluorescent protein positive oligodendrocytes ex vivo. We have identified 145 miRNAs that are expressed in oligodendrocyte cell lineage progression. Dicer1 expression decreases in differentiated oligodendrocytes, and knock down of Dicer1 results in changes in miRNAs similar to those associated with differentiation. To identify miRNAs that control the PLP expression, we have selected miRNAs whose expression is lower in differentiated vs. undifferentiated Oli-neu cells and that have one or more binding site(s) in the PLP 3'-untranslated region (3'UTR). The PLP 3'UTR fused to the luciferase gene reduces the activity of the reporter, suggesting that it negatively regulates message stability or translation. Such suppression is relieved by knock down of miR-20a. Overexpression of miR-20a decreases expression of the endogenous PLP in primary oligodendrocytes and of the reporter gene. Deletion or mutation of the putative binding site for miR-20a in the PLP 3'UTR abrogated such effects. Our data indicate that miRNA expression is regulated by Dicer1 levels in differentiated oligodendrocytes and that miR-20a, a component of the cluster that controls oligodendrocyte cell number, regulates PLP gene expression through its 3'UTR.


Assuntos
RNA Helicases DEAD-box/genética , Regulação da Expressão Gênica/genética , MicroRNAs/genética , Proteína Proteolipídica de Mielina/biossíntese , Oligodendroglia/metabolismo , Ribonuclease III/genética , Animais , Western Blotting , Diferenciação Celular/genética , Separação Celular , RNA Helicases DEAD-box/metabolismo , Camundongos , MicroRNAs/metabolismo , Proteína Proteolipídica de Mielina/genética , Oligodendroglia/citologia , Reação em Cadeia da Polimerase , Ribonuclease III/metabolismo , Transcriptoma , Transfecção
6.
Neurobiol Dis ; 44(2): 231-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21784154

RESUMO

Pelizaeus-Merzbacher disease (PMD) most frequently results from duplication of the Plp1 gene with a correlation between disease severity and increasing copy number of the gene. Animal models of PMD, in particular those overexpressing the Plp1 gene, have been sought in attempts to provide systems in which potential therapies can be tested. Here we describe a rat model of the severe connatal form of PMD and provide a detailed characterization of its pathology and molecular biology, prior to testing therapeutic approaches. We determined the exact copy number of Plp1, and the resulting effects on RNA and protein expression. Distinct differences in myelin and disparate distributions of myelin protein markers in comparison to wild-type controls were observed. Altered expression of Plp1 also caused an increase in the apoptotic cell death of oligodendrocytes. These results provide the platform from which to test the effectiveness of in vivo therapies.


Assuntos
Predisposição Genética para Doença/genética , Proteína Proteolipídica de Mielina/genética , Bainha de Mielina/genética , Bainha de Mielina/patologia , Doença de Pelizaeus-Merzbacher/genética , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Modelos Animais de Doenças , Humanos , Proteína Proteolipídica de Mielina/biossíntese , Doença de Pelizaeus-Merzbacher/patologia , Ratos , Ratos Endogâmicos Lew , Ratos Transgênicos , Ratos Wistar
7.
Cancer Res ; 71(13): 4686-95, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21551250

RESUMO

Stem cells are under strict regulation by both intrinsic factors and the microenvironment. There is increasing evidence that many cancers initiate through acquisition of genetic mutations (loss of intrinsic control) in stem cells or their progenitors, followed by alterations of the surrounding microenvironment (loss of extrinsic control). In neurofibromatosis type 1 (NF1), deregulation of Ras signaling results in development of multiple neurofibromas, complex tumors of the peripheral nerves. Neurofibromas arise from the Schwann cell lineage following loss of function at the NF1 locus, which initiates a cascade of interactions with other cell types in the microenvironment and additional cell autonomous modifications. In this study, we sought to identify whether a temporal "window of opportunity" exists during which cells of the Schwann cell lineage can give rise to neurofibromas following loss of NF1. We showed that acute loss of NF1 in both embryonic and adult Schwann cells can lead to neurofibroma formation. However, the embryonic period when Schwann cell precursors and immature Schwann cells are most abundant coincides with enhanced susceptibility to plexiform neurofibroma tumorigenesis. This model has important implications for understanding early cellular events that dictate neurofibroma development, as well as for the development of novel therapies targeting these tumors.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Genes da Neurofibromatose 1 , Neurofibroma Plexiforme/genética , Neurofibroma Plexiforme/patologia , Células de Schwann/fisiologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hiperplasia , Integrases/biossíntese , Integrases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/biossíntese , Proteína Proteolipídica de Mielina/genética , Nervos Periféricos/patologia , Gravidez , Recombinação Genética , Células de Schwann/patologia , Tamoxifeno/farmacologia , Transgenes
8.
Cancer Res ; 71(13): 4675-85, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21551249

RESUMO

Plexiform neurofibromas are peripheral nerve sheath tumors initiated by biallelic mutation of the NF1 tumor suppressor gene in the Schwann cell lineage. To understand whether neurofibroma formation is possible after birth, we induced Nf1 loss of function with an inducible proteolipid protein Cre allele. Perinatal loss of Nf1 resulted in the development of small plexiform neurofibromas late in life, whereas loss in adulthood caused large plexiform neurofibromas and morbidity beginning 4 months after onset of Nf1 loss. A conditional EGFP reporter allele identified cells showing recombination, including peripheral ganglia satellite cells, peripheral nerve S100ß+ myelinating Schwann cells, and peripheral nerve p75+ cells. Neurofibromas contained cells with Remak bundle disruption but no recombination within GFAP+ nonmyelinating Schwann cells. Extramedullary lympho-hematopoietic expansion was also observed in PlpCre;Nf1fl/fl mice. These tumors contained EGFP+/Sca-1+ stromal cells among EGFP-negative lympho-hematopoietic cells indicating a noncell autonomous effect and unveiling a role of Nf1-deleted microenvironment on lympho-hematopoietic proliferation in vivo. Together these findings define a tumor suppressor role for Nf1 in the adult and narrow the range of potential neurofibroma-initiating cell populations.


Assuntos
Inativação Gênica , Genes da Neurofibromatose 1 , Integrases/biossíntese , Proteína Proteolipídica de Mielina/biossíntese , Neurofibromatose 1/genética , Tamoxifeno/farmacologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Integrases/genética , Masculino , Camundongos , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/genética
9.
J Neurosci Res ; 89(7): 1070-8, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21472765

RESUMO

The use of immortalized cells has been instrumental as a tool with which to study gene regulation. However, it is crucial to understand the status of a given cell line, especially when investigating the regulation of genes whose expression is developmentally regulated. Several immortalized cell lines have been derived from primary cultures of mouse oligodendrocytes. Two such cell lines, N20.1 and Oli-neu, were characterized here in terms of their relative expression of myelin genes at both the mRNA level and the protein level. Analysis of the splice isoforms expressed by the myelin proteolipid protein (Plp1), myelin basic protein (Mbp), and 2',3'-cyclic nucleotide 3'-phosphodiesterase (Cnp) genes, along with the relative amount of protein expressed by these genes, suggests that the cell lines are representative of immature oligodendrocytes, although Oli-neu cells appear to be farther along the differentiation pathway compared with N20.1 cells. Previous studies have shown that the developmental increase in Plp1 gene expression that occurs during the active myelination period is governed by transcription regulatory elements present within the first intron. The responsiveness of one of these elements, the so-called antisilencer/enhancer (ASE), was investigated in both cell lines. Results presented here suggest that the ASE has a much more potent effect in Oli-neu cells. Thus, the two cell lines appear to be at different stages and will be useful as a means to study transcription regulatory elements whose influence changes during development.


Assuntos
Proteína Básica da Mielina/genética , Proteína Proteolipídica de Mielina/genética , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Diester Fosfórico Hidrolases/genética , 2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase , Animais , Diferenciação Celular/genética , Linhagem Celular Transformada , Linhagem da Célula/genética , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Camundongos , Proteína Básica da Mielina/biossíntese , Proteína Proteolipídica de Mielina/biossíntese , Oligodendroglia/enzimologia , Diester Fosfórico Hidrolases/biossíntese , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética
10.
J Neurosci ; 30(42): 13943-54, 2010 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-20962216

RESUMO

Myelinated fibers are organized into specialized domains that ensure the rapid propagation of action potentials and are characterized by protein complexes underlying axoglial interactions. TAG-1 (Transient Axonal Glycoprotein-1), a cell adhesion molecule of the Ig superfamily, is expressed by neurons as well as by myelinating glia. It is essential for the molecular organization of myelinated fibers as it maintains the integrity of the juxtaparanodal region through its interactions with Caspr2 and the voltage-gated potassium channels (VGKCs) on the axolemma. Since TAG-1 is the only known component of the juxtaparanodal complex expressed by the glial cell, it is important to clarify its role in the molecular organization of juxtaparanodes. For this purpose, we generated transgenic mice that exclusively express TAG-1 in oligodendrocytes and lack endogenous gene expression (Tag-1(-/-);plp(Tg(rTag-1))). Phenotypic analysis clearly demonstrates that glial TAG-1 is sufficient for the proper organization and maintenance of the juxtaparanodal domain in the CNS. Biochemical analysis shows that glial TAG-1 physically interacts with Caspr2 and VGKCs. Ultrastructural and behavioral analysis of Tag-1(-/-);plp(Tg(rTag-1)) mice shows that the expression of glial TAG-1 is sufficient to restore the axonal and myelin deficits as well as the behavioral defects observed in Tag-1(-/-) animals. Together, these data highlight the pivotal role of myelinating glia on axonal domain differentiation and organization.


Assuntos
Moléculas de Adesão Celular Neuronais/biossíntese , Moléculas de Adesão Celular Neuronais/genética , Mutação/genética , Mutação/fisiologia , Neuroglia/metabolismo , Neuroglia/ultraestrutura , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Comportamento Animal/fisiologia , Western Blotting , Células Cultivadas , Sistema Nervoso Central/citologia , Sistema Nervoso Central/crescimento & desenvolvimento , Contactina 2 , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Eletrônica , Proteína Proteolipídica de Mielina/biossíntese , Proteína Proteolipídica de Mielina/genética , Bainha de Mielina/genética , Bainha de Mielina/fisiologia , Oligodendroglia/metabolismo , Nervo Óptico/citologia , Nervo Óptico/crescimento & desenvolvimento , Nervo Óptico/fisiologia , Doenças do Nervo Óptico/genética , Doenças do Nervo Óptico/patologia , Equilíbrio Postural/fisiologia , Regiões Promotoras Genéticas , Células de Schwann/fisiologia
11.
ASN Neuro ; 2(4): e00043, 2010 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-20885931

RESUMO

PMD (Pelizaeus-Merzbacher disease) is a rare neurodegenerative disorder that impairs motor and cognitive functions and is associated with a shortened lifespan. The cause of PMD is mutations of the PLP1 [proteolipid protein 1 gene (human)] gene. Transgenic mice with increased Plp1 [proteolipid protein 1 gene (non-human)] copy number model most aspects of PMD patients with duplications. Hypomyelination and demyelination are believed to cause the neurological abnormalities in mammals with PLP1 duplications. We show, for the first time, intense microglial reactivity throughout the grey and white matter of a transgenic mouse line with increased copy number of the native Plp1 gene. Activated microglia in the white and grey matter of transgenic mice are found as early as postnatal day 7, before myelin commences in normal cerebra. This finding indicates that degeneration of myelin does not cause the microglial response. Microglial numbers are doubled due to in situ proliferation. Compared with the jp (jimpy) mouse, which has much more oligodendrocyte death and hardly any myelin, microglia in the overexpressors show a more dramatic microglial reactivity than jp, especially in the grey matter. Predictably, many classical markers of an inflammatory response, including TNF-α (tumour necrosis factor-α) and IL-6, are significantly up-regulated manyfold. Because inflammation is believed to contribute to axonal degeneration in multiple sclerosis and other neurodegenerative diseases, inflammation in mammals with increased Plp1 gene dosage may also contribute to axonal degeneration described in patients and rodents with PLP1 increased gene dosage.


Assuntos
Química Encefálica/genética , Regulação da Expressão Gênica , Mediadores da Inflamação/fisiologia , Microglia/metabolismo , Microglia/patologia , Proteína Proteolipídica de Mielina/biossíntese , Proteína Proteolipídica de Mielina/genética , Regulação para Cima/genética , Animais , Feminino , Dosagem de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Jimpy , Camundongos Mutantes Neurológicos , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/fisiologia
12.
Glia ; 58(14): 1727-38, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20629189

RESUMO

The most common cause of Pelizaeus-Merzbacher (PMD) is due to duplication of the PLP1 gene but it is unclear how increased gene dosage affects PLP turnover and causes dysmyelination. We have studied the dynamics of PLP/DM20 in a transgenic mouse model of PMD with increased gene dosage of the proteolipid protein gene (Plp1). The turnover of PLP/DM20 were investigated using an ex-vivo brain slice system and cultured oligodendrocytes. Homozygous mice have reduced PLP translation, markedly enhanced PLP degradation, and markedly reduced incorporation of PLP into myelin. Proteasome inhibition (MG132) prevented the enhanced degradation. Numerous autophagic vesicles are present in homozygous transgenic mice that may influence protein dynamics. Surprisingly, promoting autophagy with rapamycin decreases the degradation of nascent PLP suggesting autophagic vacuoles serve as a cellular storage compartment. We suggest that there are multiple subcellular fates of PLP/DM20 when overexpressed: the vast majority being degraded by the proteasome, a proportion sequestered into autophagic vacuoles, probably fused with endolysosomes, and only a small proportion entering the myelin sheath, where its association with lipid rafts is perturbed. Transgenic oligodendrocytes have fewer membrane sheets and this phenotype is improved with siRNA-mediated knockdown of PLP expression that promotes the formation of MBP+ myelin-like sheets. This finding suggests that RNAi technology is in principle applicable to improve CNS myelination when compromised by PLP/DM20 overexpression.


Assuntos
Predisposição Genética para Doença/genética , Proteína Proteolipídica de Mielina/genética , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Doença de Pelizaeus-Merzbacher/genética , Doença de Pelizaeus-Merzbacher/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/antagonistas & inibidores , Proteína Proteolipídica de Mielina/biossíntese , Técnicas de Cultura de Órgãos , Interferência de RNA/fisiologia , Vacúolos/metabolismo , Vacúolos/ultraestrutura
13.
J Neurosci Res ; 87(13): 2842-53, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19479988

RESUMO

The proteolipid protein (PLP) gene (Plp) encodes the major myelin proteins, PLP and DM20. Expression of Plp occurs predominantly in oligodendrocytes, but evidence is accumulating that this gene is also expressed in neurons. In earlier studies, we demonstrated that myelin-deficient (MD) rats, which carry a mutation in the Plp gene, exhibit lethal hypoxic ventilatory depression. Furthermore, we found that, in the MD rat, PLP accumulated in neuronal cell bodies in the medulla oblongata. In the current study, we sought to determine which neurons expressed the Plp gene in the medulla oblongata and whether Plp gene expression changed in neurons with maturation. A transgenic mouse expressing the Plp promoter driving expression of enhanced green fluorescent protein (Plp-EGFP) was used to identify neurons expressing this gene. Plp expression in neurons was confirmed by immunostaining EGFP-positive cells for NeuN and by in situ hybridization for PLP mRNA. The numbers of neurons expressing Plp-EGFP and their distribution increased between P5 and P10 in the medulla. Immunostaining for surface receptors and classes of neurons expressing Plp-EGFP revealed that Plp gene expression in brainstem neurons was restricted to neurons expressing specific ligand-gated channels and biosynthetic enzymes, including glutamatergic NMDA receptors, GABA(A) receptors, and ChAT in defined areas of the medulla. Plp gene expression was rarely found in interneurons expressing GABA and was never found in AMPA receptor- or tyrosine hydroxylase-expressing neurons. Thus, Plp expression in the mouse caudal medulla was found to be developmentally regulated and restricted to specific groups of neurons.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Bulbo/metabolismo , Proteína Proteolipídica de Mielina/biossíntese , Neurônios/metabolismo , Animais , Diferenciação Celular , Colina O-Acetiltransferase/análise , Feminino , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Masculino , Bulbo/citologia , Bulbo/crescimento & desenvolvimento , Camundongos , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/genética , Proteínas do Tecido Nervoso/análise , Neurônios/química , Regiões Promotoras Genéticas , Receptores de AMPA/análise , Receptores de GABA-A/análise , Receptores de Glutamato Metabotrópico/análise , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/biossíntese , Tirosina 3-Mono-Oxigenase/análise , Ácido gama-Aminobutírico/análise
14.
Mol Cell Neurosci ; 38(4): 489-94, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18555697

RESUMO

We have recently reported that overexpression of proteolipid protein in oligodendrocytes leads to a pathologically relevant increase of both CD8+ T-lymphocytes and CD11b+ cells in the CNS. We now focussed on the origin of the CD11b+ cells in the optic nerve, a well established structure for the analysis of the mutant, using bone marrow chimeric mice. Although there is an age-related increase in CD11b+ cells in the myelinated part of the optic nerve of the mutants, the percentage of infiltrating cells was not increased, but enhanced proliferation was detectable. In the non-myelinated optic nerve head, the rate of infiltrating CD11b+ cells and albumin extravasation was high in both genotypes. However, albumin extravasation was also high in the rostral myelinated part, where CD11b+ cell influx was low. Our study demonstrates an intrinsic origin of CD11b+ cells in the presence of an unchanged blood-brain-barrier in a CNS myelin mutant.


Assuntos
Barreira Hematoencefálica/patologia , Antígeno CD11b/biossíntese , Diferenciação Celular/genética , Macrófagos/patologia , Proteína Proteolipídica de Mielina/genética , Nervo Óptico/patologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Transplante de Medula Óssea/métodos , Antígeno CD11b/genética , Diferenciação Celular/imunologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/biossíntese , Nervo Óptico/imunologia , Nervo Óptico/metabolismo
15.
Neurochem Res ; 32(2): 377-88, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17203404

RESUMO

Irradiation is one way to condition Twitcher mice--a natural model of globoid cell leukodystrophy (GLD)--prior to receive bone marrow transplantation (BMT). BMT showed to delay but not to completely prevent GLD disease in treated mutants. The reasons why BMT is not completely preventive in Twitchers are unclear but we speculate that irradiation might contribute to worsen the neurological impairments generated by the disease by altering postnatal neurogenesis. To test this hypothesis, we examined proliferation, migration and differentiation of neural precursors in neurogenic areas of the Twitcher brain after exposure of 5 day-old mutant pups to 620 rad, a non-lethal dose that leads to 80-90% of bone-marrow engraftment in classic BMT. Twitchers showed to be sensitive to irradiation, leading to a severe retardation of body growth of irradiated mutants. Irradiated Twitchers had reduced proliferation of neural precursors and increased astrogliosis and microgliosis, with reduced numbers of migratory neuroblasts and significantly less brain myelination. These effects were accompanied by caspase-3 activation and appeared largely irreversible in the lifespan of the Twitcher. Our work confirms that exposure of the neonatal brain to irradiation conditions such as those performed prior to BMT, can lead to long-lasting alterations of postnatal neurogenesis and myelination, which might contribute to worsen the progression of disease in these myelin mutants and to reduce the success of BMT.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/efeitos da radiação , Raios gama , Leucodistrofia de Células Globoides/fisiopatologia , Animais , Apoptose/efeitos da radiação , Transplante de Medula Óssea , Encéfalo/citologia , Caspase 3/metabolismo , Proliferação de Células/efeitos da radiação , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Macrófagos/efeitos da radiação , Camundongos , Camundongos Mutantes Neurológicos , Proteína Proteolipídica de Mielina/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Neuroglia/fisiologia , Neuroglia/efeitos da radiação , Neurônios/fisiologia , Neurônios/efeitos da radiação
16.
Neurochem Res ; 32(2): 293-302, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17235686

RESUMO

Despite a substantial understanding of the factors regulating oligodendrocyte differentiation, the signaling mechanisms involved in this process are not well-understood. This study elaborates on the findings (Bhat NR, Zhang P (1997) FASEB J 11:A925; Baron W, Metz B, Bansal R, Hoekstra D, de Vries H (2000) Mol Cell Neurosci 15:314-329) of a role for p38 MAP kinase signaling in oligodendrocyte differentiation and myelin gene expression. When proliferating oligodendrocyte progenitors were switched to a growth factor-free differentiation medium, there was a rapid activation of p38 kinase that correlated with an increased phosphorylation of CREB, a down-stream target and a factor involved in oligodendrocyte differentiation. Addition of forskolin, a known inducer of intracellular c-AMP and of oligodendrocyte differentiation, also stimulated CREB phosphorylation in a p38 kinase dependent way. Pharmacological inhibition of p38 interfered with the morphological and antigenic changes associated with differentiating oligodendrocytes as well as with the developmental and forskolin-induced expression of myelin basic protein, thereby supporting an essential role for p38 MAPK pathway in oligodendrocyte differentiation.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Oligodendroglia/citologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Colforsina/farmacologia , Ativação Enzimática , Imidazóis/farmacologia , Proteína Básica da Mielina/biossíntese , Proteína Proteolipídica de Mielina/biossíntese , Glicoproteína Associada a Mielina/biossíntese , Oligodendroglia/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Piridinas/farmacologia , RNA Mensageiro/antagonistas & inibidores , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
17.
Neurochem Res ; 32(2): 343-51, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17191136

RESUMO

Jimpy (Plp(jp)) is an X-linked recessive mutation in mice that causes CNS dysmyelination and early death in affected males. It results from a point mutation in the acceptor splice site of myelin proteolipid protein (Plp) exon 5, producing transcripts that are missing exon 5, with a concomitant shift in the downstream reading frame. Expression of the mutant PLP product in Plp(jp) males leads to hypomyelination and oligodendrocyte death. Expression of our Plp-lacZ fusion gene, PLP(+)Z, in transgenic mice is an excellent readout for endogenous Plp transcriptional activity. The current studies assess expression of the PLP(+)Z transgene in the Plp(jp) background. These studies demonstrate that expression of the transgene is decreased in both the central and peripheral nervous systems of affected Plp(jp) males. Thus, expression of mutated PLP protein downregulates Plp gene activity both in oligodendrocytes, which eventually die, and in Schwann cells, which are apparently unaffected in Plp(jp) mice.


Assuntos
Sistema Nervoso Central/metabolismo , Proteína Proteolipídica de Mielina/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Sistema Nervoso Periférico/metabolismo , Animais , Western Blotting , Sistema Nervoso Central/crescimento & desenvolvimento , Regulação para Baixo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Óperon Lac/genética , Masculino , Camundongos , Camundongos Jimpy , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/genética , Proteínas do Tecido Nervoso/genética , Oligodendroglia/metabolismo , Células de Schwann/metabolismo , Transgenes/genética , beta-Galactosidase/biossíntese
18.
Glia ; 55(4): 341-51, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17133418

RESUMO

Duplication of PLP1, an X-linked gene encoding the major myelin membrane protein of the human CNS, is the most frequent cause of Pelizaeus-Merzbacher disease (PMD). Transgenic mice with extra copies of the wild type Plp1 gene, a valid model of PMD, also develop a dysmyelinating phenotype dependant on gene dosage. In this study we have examined the effect of increasing Plp1 gene dosage on levels of PLP/DM20 and on other representative myelin proteins. In cultured oligodendrocytes and early myelinating oligodendrocytes in vivo, increased gene dosage leads to elevated levels of PLP/DM20 in the cell body. During myelination, small increases in Plp1 gene dosage (mice hemizygous for the transgene) elevate the level of PLP/DM20 in oligodendrocyte soma but cause only minimal and transient effects on the protein composition and structure of myelin suggesting that cells can regulate the incorporation of proteins into myelin. However, larger increases in dosage (mice homozygous for the transgene) are not well tolerated, leading to hypomyelination and alteration in the cellular distribution of PLP/DM20. A disproportionate amount of PLP/DM20 is retained in the cell soma, probably in autophagic vacuoles and lysosomes whereas the level in myelin is reduced. Increased Plp1 gene dosage affects other myelin proteins, particularly MBP, which is transitorily reduced in hemizygous mice but consistently and markedly lower in homozygotes in both myelin and naïve or early myelinating oligodendrocytes. Whether the reduced MBP is implicated in the pathogenesis of dysmyelination is yet to be established.


Assuntos
Proteínas da Mielina/biossíntese , Proteína Proteolipídica de Mielina/biossíntese , Bainha de Mielina/metabolismo , Proteínas do Tecido Nervoso/genética , Doença de Pelizaeus-Merzbacher/metabolismo , Animais , Northern Blotting , Western Blotting , Contagem de Células , Células Cultivadas , Dosagem de Genes , Expressão Gênica/fisiologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/genética , Oligodendroglia/metabolismo , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/metabolismo
19.
J Neurosci Res ; 84(6): 1206-16, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16917838

RESUMO

Myelin proteolipid protein (plp), a major myelin protein in the CNS, has been proposed to function in myelin assembly. Transgenic mice overexpressing the plp gene by introduction of two extra wild-type (Wt) mouse plp genes (plp(tg/-)) exhibit normal myelination and ion channel clustering at the age of 2 months. However, at the age of 5 months, demyelination becomes observable, accompanied by a reduction in the number of K+ channel clusters at Ranvier's node and a progressive increase in motor deficit. To clarify how these age-dependent changes are related to nerve conduction in the CNS, we analyzed the conduction velocity (CV) and relative refractory period (RRP) of identified spinal ascending or descending tracts, such as the dorsal column pathway, the vestibulospinal and reticulospinal tracts, and the pyramidal tract, in plp(tg/-) mice 2, 5, and 8 months of age. We found that CVs decreased as age increased. Importantly, CVs were significantly reduced and prolonged RRPs were observed in 2-month-old (2M) plp(tg/-) mice that had no apparent demyelination. Immunohistological examination revealed that densities of Na+ and K+ channel clusters decreased as plp(tg/-) and Wt mice aged. However, a clear correlation was not observed between CVs and mean channel cluster densities or between mean channel cluster densities and progress of demyelination. Performance in the rotarod test was normal in 2M plp(tg/-) mice but deteriorated in mice older than age 5 months. These results suggest that electrophysiological analysis can detect the abnormalities of the plp(tg/-) mice earlier than histological or behavioral measures.


Assuntos
Sistema Nervoso Central/fisiopatologia , Doenças Desmielinizantes/fisiopatologia , Proteína Proteolipídica de Mielina/biossíntese , Proteína Proteolipídica de Mielina/genética , Animais , Axônios/fisiologia , Eletrofisiologia , Regulação da Expressão Gênica/fisiologia , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Transtornos dos Movimentos/genética , Condução Nervosa/fisiologia , Equilíbrio Postural/fisiologia , Canais de Potássio/fisiologia , Período Refratário Eletrofisiológico/fisiologia , Canais de Sódio/fisiologia , Medula Espinal/fisiologia
20.
Glia ; 53(7): 715-22, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16506223

RESUMO

The rumpshaker mutation of the X-linked myelin proteolipid protein (PLP1) gene causes spastic paraplegia type 2 or a mild form of Pelizaeus-Merzbacher disease in man. The identical mutation occurs spontaneously in mice. Both human and murine diseases are associated with dysmyelination. Using the mouse model, we show that the low steady state levels of PLP result from accelerated proteasomal degradation rather than decreased synthesis. The T(1/2) for degradation of rumpshaker PLP is 11 h compared with 23 h for wild type. A minority of newly synthesized PLP is incorporated into myelin in the correct orientation but at a reduced rate compared with wild type. However, inhibition of proteasomal degradation does not increase the level of PLP incorporated into myelin. As Plp null mice do not have a similar myelin deficiency, it is unlikely that the reduced PLP levels are the main cause of the dysmyelination. Rumpshaker oligodendrocytes also have a reduced level of other myelin proteins, such as MBP, although the mechanisms are not yet defined but are likely to operate at a translational or post-translational level.


Assuntos
Sistema Nervoso Central/metabolismo , Proteína Proteolipídica de Mielina/biossíntese , Proteína Proteolipídica de Mielina/genética , Bainha de Mielina/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Doença de Pelizaeus-Merzbacher/genética , Doença de Pelizaeus-Merzbacher/metabolismo , Animais , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/fisiopatologia , Modelos Animais de Doenças , Regulação para Baixo/genética , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Camundongos Mutantes Neurológicos , Mutação/genética , Bainha de Mielina/genética , Bainha de Mielina/patologia , Oligodendroglia/metabolismo , Doença de Pelizaeus-Merzbacher/fisiopatologia , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional/genética , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...